IL176765A - Inhibitors of glutaminyl cyclase - Google Patents

Inhibitors of glutaminyl cyclase

Info

Publication number
IL176765A
IL176765A IL176765A IL17676506A IL176765A IL 176765 A IL176765 A IL 176765A IL 176765 A IL176765 A IL 176765A IL 17676506 A IL17676506 A IL 17676506A IL 176765 A IL176765 A IL 176765A
Authority
IL
Israel
Prior art keywords
phenyl
inhibitors
formula
inhibitor
compound
Prior art date
Application number
IL176765A
Other versions
IL176765A0 (en
Original Assignee
Probiodrug Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/838,993 external-priority patent/US7371871B2/en
Priority claimed from PCT/EP2005/001153 external-priority patent/WO2005075436A2/en
Application filed by Probiodrug Ag filed Critical Probiodrug Ag
Publication of IL176765A0 publication Critical patent/IL176765A0/en
Publication of IL176765A publication Critical patent/IL176765A/en

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

176765 ρ'Π I 453530 τηχ Novel inhibitors of glutaminyl cyclase Probiodrug AG C. 168243 Novel Inhibitors of Glutaminyl Cyclase Field of the invention The invention relates to glutaminyl cyclase (QC, EC 2.3.2.5) that 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 Canca 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 (Bookers, 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, 11246-11250), 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 Glu-i-conversion is favored around pH 6.0 while Glnrconversion to pGlu-derivatives occurs with a pH-optimum of around 8.0. Since the formation of pGlu-Ap-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.
EP 02 011 349.4 discloses polynucleotides encoding insect glutaminyl cyclase, as well as polypeptides encoded thereby. This application further provides host cells comprising expression vectors comprising polynucleotides of the invention. Isolated polypeptides and host cells comprising insect QC are useful in methods of screening for agents that reduce glutaminyl cyclase activity. Such agents are useful as pesticides.
Definitions Enzyme inhibitors Reversible enzyme inhibitors: comprise competitive inhibitors, non-competitive reversible inhibitors, slow-binding or tight-binding inhibitors, transition state analogs and multisubstrate analogs.
Competitive inhibitors show i) non-covalent interactions with the enzyme, ii) compete with substrate for the enzyme active site, The principal mechanism of action of a reversible enzyme inhibitor and the definition of the dissociation constant can be visualized as follows: E-S E-P c: r E ♦ P The formation of the enzyme-inhibitor [E-l] complex prevents binding of substrates, therefore the reaction cannot proceed to the normal physiological product, P. A larger inhibitor concentration [I] leads to larger [E-l], leaving less free enzyme to which the substrate can bind.
Non-competitive reversible inhibitors i) bind at a site other than active site (allosteric binding site) ii) cause a conformational change in the enzyme which decreases or stops catalytic activity.
Slow-binding or tight-binding inhibitors i) are competitive inhibitors where the equilibrium between inhibitor and enzyme is reached slowly, ii) (kon is slow), possibly due to conformational changes that must occur in the enzyme or inhibitor a) are often transition state analogs b) are effective at concentrations similar to the enzyme cone, (subnanomolar KD values) C) due to koff values being so low these types of inhibitors are "almost" irreversible Transition state analogs are competitive inhibitors which mimic the transition state of an enzyme catalyzed reaction. Enzyme catalysis occurs due to a lowering of the energy of the transition state, therefore, transition state binding is favored over substrate binding.
Multisubstrate Analogs For a reaction involving two or more substrates, a competitive inhibitor or transition state analog can be designed which contains structural characteristics resembling two or more of the substrates.
Irreversible enzyme inhibitors: drive the equilibrium between the unbound enzyme and inhibitor and enzyme inhibitor complex (E + I <— > E-J) all the way to the right with a covalent bond (~100 kcal/mole), making the inhibition irreversible.
Affinity labeling agents • Active-site directed irreversible inhibitors (competitive irreversible inhibitor) are recognized by the enzyme (reversible, specific binding) followed by covalent bond formation, and i) are structurally similar to substrate, transition state or product allowing for specific interaction between drug and target enzyme, contain reactive functional group (e.g. a nucleophile, -COCH2Br) allowing for covalent bond formation The reaction scheme below describes an active-site directed reagent with its target enzyme where KD is the dissociation constant and ηΒίΛ1ν3«οη is the rate of covalent bond formation.
E+ / < Ko > £ » / nyB"eK ) E- I • Mechanism-based enzvme inactivators (also called suicide inhibitors) are active-site directed reagents (unreactive) which binds to the enzyme active site where it is transformed to a reactive form (activated) by the enzyme's catalytic capabilities. Once activated, a covalent bond between the inhibitor and the enzyme is formed.
The reaction scheme below shows the mechanism of action of a mechanism based enzyme inactivator, where KD is the dissociation complex, k2 is the rate of activation of the inhibitor once bound to the enzyme, k3 is the rate of dissociation of the activated inhibitor, P, from the enzyme (product can still be reactive) from the enzyme and k, is the rate of covalent bond formation between the activated inhibitor and the enzyme.
Inactivation (covalent bond formation, k4) must occur prior to dissociation (k3) otherwise the now reactive inhibitor is released into the environment. Partition ratio, ka/k ratio of released product to inactivation should be minimized for efficient inactivation of the system and minimal undesirable side reactions. A large partition ratio (favors dissocation) leads to nonspecific reactions.
Uncompetitive enzyme inhibitors: From the definition of uncompetitive inhibitor (an inhibitor which binds only to ES complexes) the following equilibria can be written: Ks k2 E + S ES - E + P + I ESI The ES complex dissociates the subtrate with a dissociation constant equal to Ks, whereas the ESI complex does not dissociate it (i.e has a Ks value equal to zero). The Km's of Michaelis-Menten type enzymes are expected to be reduced. Increasing substrate concentration leads to increasing ESI concentration (a complex incapable of progressing to reaction products), therefore the inhibition can not be removed.
Preferred according to the present invention are competitive enzyme inhibitors.
Most preferred are competitive reversible enzyme inhibitors.
The terms "kf or "Κ ' and "KDn are binding constants, which describe the binding of an inhibitor to and the subsequent release from an enzyme. Another measure is the "IC50" value, which reflects the inhibitor concentration, which at a given substrate concentration results in 50 % enzyme activity.
The term "DP IV-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 activityof 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 were 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.
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-p-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 side 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 a Ki 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 0.01 μΜ or less. Indeed, inhibitors with Ki 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 convience, 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 1000 g/mole or less, 500 g/mole or less, preferably of 400 g/mole or less, and even more preferably of 350 g/mole or less and even of 300 g/mole or less.
The term "subjecf 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 "acyl", unless specifically limited, denotes a C1.12 acyl residue, preferably a C1-3 acyl residue and especially preferred a C1- acyl residue. Examples of acyl include alkanoyl groups mentioned below and benzoyl.
"Peptides" are selected from dipeptides to decapeptides, preferred are dipeptides, tripeptides, tetrapeptides and pentapeptides. The amino acids for the formation of the "peptides" can be selected from those listed below.
Throughout the description and the claims the expression "alkyl", unless specifically limited, denotes a C1-12 alkyl group, preferably a C1-6 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), (n-butyl, ferf-butyl and sec-butyl), pentyl, hexyl, heptyl (e.g. n-heptyl) and octyl (e.g. n-octyl). The expression "alk", for example in the expression "alkoxy", and the expression "alkan", for example in the expression "alkanoyl", should be interpreted in accordance with the definition of "alkyl". Exemplary alkoxy groups include methoxy, ethoxy, butoxy (e.g. n-butoxy), heptyloxy (e.g. n-heptyloxy) and octyloxy (e.g. n-octyloxy). Exemplary alkanoyl (i.e. acyl groups) include ethanoyl (i.e. acetyl), propionyl and butyryl.
The expression "alkenyl", unless specifically limited, denotes a C2-12 alkenyl group, preferably a C2-6 alkenyl group, which contains at least one double bond at any desired location. Alkenyl groups may be straight chain or branched. Exemplary alkenyl groups include ethenyl, propenyl and butenyl.
The expression "alkynyl", unless specifically limited, denotes a C2-12 alkynyl group, preferably a C2-6 alkynyl group, which contains at least one triple bond at any desired location. Alkynyl groups may be straight chain or branched. Exemplary alkenyl groups include ethynyl, propynyl and butynyl.
The expression "cycloalkyl", unless specifically limited, denotes a C3-12 cycloalkyl group, preferably a C3.8 cycloalkyl group. Exemplary cycloalkyl groups include cylcopropyl, cyclobutyl, cyclopropyl, cyclohexyl, cycloheptyl and cyclooctyl. Cycloalkyl groups may be branched in which case the number of carbons indicates the total number of carbons in the moiety.
The expression "heterocyclic", unless specifically limited, denotes a cycloalkyl residue, wherein one or more (e.g. 1, 2 or 3) ring atoms are replaced by heteroatoms selected from N, S or O. Exemplary heterocyclic groups containing one hetero atom include pyrrolidine, tetrahydrofuran and piperidine. Such groups may be optionally substituted eg by alkyl, oxo or hydroxyl.
Concrete examples of a heterocyclic group comprise a substituted or unsubstituted oxirano, aziridino, oxacyclopropyl, azacyclopropyl, thiirano, oxetano, thietano, pyrrolidine tetrahydrofurano, thioiano, 1,1-dioxo-thiolano, 1,3-dioxolano, thiazolidino, imidazolidino, oxazolidino, pyrazolidino, tetrahydropyrano, piperidino, urotropino, piperazino, N-methyl-piperazino, (2-(N-methyl)-N'-piperazinyl)-ethyl, (4Ν-(2'-hydroxyethyl)-1 N-piperazinyl), (2-(4N-(2'-hydroxyethyl)-1 N-piperazinyl)-ethyloxy), morpholino, 2-(N-morphoiino)-ethyl group, as well as lactams, lactones, cyclic imides and cyclic anhydrides.
The expression "carbocylic", unless specifically limited, denotes a carbocylic group containing between 3 and 12 carbon atoms, more preferably between 3 and 8 carbon atoms. A carboyclic group, as used herein, refers to a group other than aryl or cycloalkyl which comprises at least one ring of carbon atoms without heteroatoms. Examples of carbocylic groups include bridged ring systems (e.g. bicyclo[2.2.1]heptenyl) and partially unsaturated ring systems.
The expression "aryl", unless specifically limited, denotes a C5-12 aryl group, preferably a Cs-s aryl group. Aryl groups will contain at least one aromatic ring (e.g. one, two or three rings), but may also comprise partially or fully unsaturated rings. An example of an aryl group with one aromatic ring is phenyl. Examples of aromatic groups with two aromatic rings include naphyl. Examples of aryl groups which contain partially or fully unsaturated rings include pentalene and indene. As noted below, aryl groups may optionally be substituted. Further examples for aryl groups are 4-fluoro-phenyl, 3-fluoro-phenyl, pentafluoro-phenyl, 4-hydroxyphenyl- 3-nitro-phenyl-, 4-(trifluoromethyl)-phenyl-, 4-anilinyl-, 2-biphenylyl-, 3-biphenylyl-, 4-biphenylyl-, indenyl-, -naphthyl-, or 2-naphthyl-, -anthracenyl-, 2-anthracenyl-, 3-anthracenyl- groups.
Examples of -alkylaryl include phenylmethyl- (benzyl) and phenylethyl, 2-phenyIeth-1-yl, ρ-tolyl-methyl-, p-tolyl-ethyl-, m-tolyl-methyl-, /77-tolyl-ethyh o-tolyl-methyl-, o-tolyl-ethyl-, 2-(4-ethyl-phenyl)-eth-1-yl-, 2,3-dimethyl-phenyl-methyl-, 2,4-dimethyl-phenyl-methyl-, 2,5-dimethyl-phenyl-methyl-, 2,6-dimethyl-phenyl-methyl-, 3,4-dimethyl-phenyl-methyl-, 3,5-dimethyl-phenyl-methyl-, 2,4,6-trimethyl-phenyl-methyl-, 2,3- dimethyl-phenyl-ethyl-, 2,4-dimethyl-phenyl-ethyl-, 2,5-dimethyl-phenyl-ethyl-, 2,6-dimethyl-phenyl-ethyl-, 3,4-dimethyl-phenyl-ethyl-, 3,5-dimethyl-phenyl-ethyl-, 2,4,6-trimethyl-phenyl-ethyl-, benzhydryl (= diphenyl-methyl), benzhydryl (= diphenyl-ethyl), trityl (= triphenyl-methyl), trityl (= triphenyl-ethyl), ct-styryl, β-styryl, cumyl, 2-ethyl-phenyl-methyl-, 3-ethyl-phenyl-methyl-, 4-ethyl-phenyl-methyl-, 2-ethyl-phenyl-ethyl-( 3-ethyl-phenyl-ethyl-, 4-ethyl-phenyl-ethyl-, 2-fluoro-benzyl, 1-methyl-2-fluoro-phen-6-yl-methyl-, 1-methyl-2-fluoro-phen-4-yl-methyl-, 1-methyl-2-fluoro-phen-6-yI-ethyl-, 1-methyl-2-fluoro-phen-4-yl-ethyl-, 1 H-indenyl-methyl-, 2H-indenyl-methyl-, 1 H-indenyl-ethyl-, 2H-indenyl-ethyl-, indanyl-methyl-, indan-1-on-2-yl-methyl-, indan-1-on-2-yl-ethyl-, tetralinyl-methyl- tetralinyl-ethyl-, fluorenyl-methyl-, fluorenyl-ethyl-, (3-phenyl)-cyclopent-1-yl ?, dihydronaphthalinyl-methyl-, dihydronaphthalinyl-ethyl-, or (4-cyclohexyl)-phenyl-methyl-, (4-cyclohexyl)-phenyl-ethyl-.
The expression "heteroary , unless specifically limited, denotes as an aryl residue, wherein one or more (e.g. 1 , 2, 3, or 4, preferably 1 , 2 or 3) ring atoms are replaced by heteroatoms selected from N, S and O or else a 5-membered aromatic ring containing one or more (e.g. 1 , 2, 3, or 4, preferably 1 , 2 or 3) ring atoms selected from N, S and O. As noted below, heteroaryl groups may optionally be substituted. Exemplary heteroaryl groups include, pyridine (eg 2, 3 or 4-pyridine), pyrimidine, quinoline, pyrrole, furan, thiophene, oxazole, pyrazole, benzodioxolane, benzodioxane, benzothiophene, benzodioxepine, and thiazolyl, 1-imidazolyl, 2-imidazolyl, 4-imidazolyl, 3-phenyl-1-pyrrolyl, isoxazolyl, isothiazolyl, 3-pyrazolyl, 1 ,2,3-triazolyl, 1 ,2,4- triazolyl, tetrazolyl, pyridazinyl, pyrazinyl, indazolyl, 6-indolyl, benzimidazolyl, isochinolinyl, purinyl, carbazolinyl, acridinyl, and 2,3'-bifuryl groups.
[Examples of -alkylheteroaryl include pyridinylmethyl-, N-methyl-pyrrol-2-methyl- N-methyl-pyrrol-2-ethyl-, N-methyl-pyrrol-3-methyl-, N-methyl-pyrrol-3-ethyl-, 2-methyl-pyrrol-1 -methyl-, 2-methyl-pyrrol-1 -ethyl-, 3-methyl-pyrrol-1 -methyl-, 3-methyl-pyrrol- 1 - ethyl-, 4-pyridino-methyl-, 4-pyridino-ethyl-, 2-(thiazol-2-yl)-ethyl-, tetrahydroisochinolinyl-methyl-, tetrahydroisochinolinyl-ethyl-, 2-ethyl-indol-1 -methyl-, 2- ethyl-indol-1 -ethyl-, 3-ethyl-indol-1 -methyl-, 3-ethyl-indol-1 -ethyl-, 4-methyl-pyridin-2-methyh 4-methyl-pyridin-2-yl-ethyl-, 4-methyl-pyridin-3-methyl, 4-methyl-pyridin-3-ethyl.
The aforementioned aryl and heteroaryl groups may, where appropriate, optionally be substituted.
The expression "substitution" or "substituted" includes the substitution by one or more (e.g. 1 , 2 or 3, preferably 1 or 2) monovalent or multivalent functional groups. Suitable substituent groups include alkyl, cycloalkyl, aryl (eg phenyl), heteroaryl (eg fury!), carbocylic, heterocyclic, alkoxy, cycloalkoxy, aryloxy, heteroaryloxy, carbocyclicoxy, hetercyclicoxy, alkenyloxy, alkynyloxy, alkenyl, alkynyl, acyl, alkanoyl, alkoxyalkanoyl, alkoxyalkyl, heteroarylalkyl, arylalkyl, arylalkyloxy, heteroarylalkyloxy, nitro, -S-alkyl (e.g. methylthio) halo (e.g. fluoro, chloro, bromo and iodo), cyano, hydroxyl, -S02alkyl, -S02aryl, -S02heteroaryl, -S02cycloalkyl -S02heterocyclic, -C02H, -C02alkyl, -NH2, -NHalkyl, -N(alkyl)2 (e.g. dimethylamino), -CO-N(alkyl)2 and -CO-NH (alkyl).
Alkyl groups including derivatives such as alkoxy together with alkenyl, alkynyl and cycloalkyl groups may optionally be halogen substituted e.g. substituted by fluoro. For example, halo substituted alkyl groups include trifluromethyl and halo substituted alkoxy groups include trifluoromethoxy.
The term "halogen" comprises fluorine (-F), chlorine (-CI), bromine (-Br), and iodine (-I), respectively.
Amino acids which can be used in the present invention are L and D-amino acids, N-alkylated amino acids, N-methyl-amino acids, aza-amino acids; alio- and threo-forms of He and Thr, which can, e.g. be α-, β- or ω-amino acids, whereof a-amino acids are preferred.
Examples of amino acids are: aspartic acid (Asp), glutamic acid (Glu), arginine (Arg), lysine (Lys), histidine (His), glycine (Gly), serine (Ser), cysteine (Cys), threonine (Thr), asparagine (Asn), glutamine (Gin), tyrosine (Tyr), alanine (Ala), proline (Pro), valine (Val), isoleucine (lie), leucine (Leu), methionine (Met), phenylalanine (Phe), tryptophan (Trp), hydroxyproline (Hyp), beta-alanine (beta-Ala), 2-aminooctanoic acid (Aoa), acetidine-(2)-carboxylic acid (Ace), pipecolic acid (Pip), 3-aminopropionic acid, 4-aminobutyric acid and so forth, alpha-aminoisobutyric acid (Aib), sarcosine (Sar), ornithine (Orn), citrulline (Cit), homoarginine (Har), t-butylalanine (f-butyl-Ala), t-butylglycine (f-butyl-Gly), N-methylisoleucine (N-Melle), phenylglycine (Phg), cyclohexylalanine (Cha), norleucine (Nle), cysteic acid (Cya) and methionine sulfoxide (MSO), acetyl-Lys, modified amino acids such as phosphoryl-serine (Ser(P)), benzyl-serine (Ser(Bzl)) and phosphoryl-tyrosine (Tyr(P)), 2-aminobutyric acid (Abu), aminoethylcysteine (AECys), carboxymethylcysteine (Cmc), dehydroalanine (Dha), dehydroamino-2-butyric acid (Dhb), carboxyglutaminic acid (Gla), homoserine (Hse), hydroxyzine (Hyl), c/s-hydroxyproline (c/sHyp), frans-hydroxyproline (fransHyp), isovaline (lva), pyroglutamic acid (Pyr), norvaline (Nva), 2-aminobenzoic acid (2-Abz), 3-aminobenzoic acid (3-Abz), 4- aminobenzoic acid (4-Abz), 4-(aminomethyl)benzoic acid (Amb), 4-(aminomethyl)cyclohexanecarboxylic acid (4-Amc), Penicillamine (Pen), 2-amino-4-cyanobutyric acid (Cba), cycloalkane-carboxylic aicds. Examples of TO-amino acids are e.g.: 5-Ara (aminoraleric acid), 6-Ahx (aminohexanoic acid), 8-Aoc (aminooctanoic aicd), 9-Anc (aminovanoic aicd), 10-Adc (aminodecanoic acid), 11-Aun (aminoundecanoic acid), 12-Ado (aminododecanoic acid). Further amino acids are: indanylglycine (Igl), indoline-2-carboxylic acid (Idc), octahydroindole-2-carboxylic acid (Oic), diaminopropionic acid (Dpr), diaminobutyric acid (Dbu), naphtylalanine (1-Nal) and (2-Nal), 4-aminophenylalanine (Phe(4-NH2)), 4-benzoylphenylalanine (Bpa), diphenylalanine (Dip), 4-bromophenylalanine (Phe(4-Br)), 2-chlorophenylalanine (Phe(2-CI)), 3-chlorophenylalanine (Phe(3-CI)), 4-chlorophenylalanine (Phe(4-CI)), 3,4-chlorophenylalanine (Phe (3,4-CI2)), 3-fluorophenylalanine (Phe(3-F)), 4-fluorophenylalanine (Phe(4-F)), 3,4-fluorophenylalanine (Phe(3,4-F2)), pentafluorophenylalanine (Phe(F5)), 4-guanidinophenylalanine (Phe(4-guanidino)), homophenylalanine (hPhe), 3-jodophenylalanine (Phe(3-J)), 4-jodophenylalanine (Phe(4-J)), 4-methylphenylalanine (Phe(4-Me)), 4-nitrophenylalanine (Phe-4-N02)), biphenylalanine (Bip), 4-phosphonomethylphenylalanine (Pmp), cyclohexylglycine (Ghg), 3-pyridinylalanine (3-Pal), 4-pyridinylalanine (4-Pal), 3,4-dehydroproline (A-Pro), 4-ketoproline (Pro(4-keto)), thioproline (Thz), isonipecotic acid (Inp), 1,2,3,4,-tetrahydroisoquinolin-3-carboxylic acid (Tic), propargylglycine (Pra), 6-hydroxynorleucine (NU(6-OH)), homotyrosine (hTyr), 3-jodotyrosine (Tyr(3-J)), 3,5-dijodotyrosine (Tyr(3,5-J2)), methyltyrosine (Tyr(Me)), 2',6 -dimethyltyrosine (Dmt), 3-N02-tyrosine (Tyr(3-N02)), phosphotyrosine (Tyr(P03H2)), alkylglycine, 1-aminoindane-1-carboxylic acid, 2-aminoindane-2-carboxylic acid (Aic), 4-amino- methylpyrrol-2-carboxyIic acid (Py), 4-amino-pyrrolidine-2-carboxyiic acid (Abpc), 2-aminotetraline-2-carboxylic acid (Ate), diaminoacetic acid (Gly(NH2)), diaminobutyric acid (Dab), 1 ,3-dihydro-2H-isoinole-carboxylic acid (Disc), homocylcohexylalanine (hCha), homophenylalanine (hPhe or Hot), frans-3-phenyl-azetidine-2-carboxylic acid, 4-phenyl-pyrroiidine-2-carboxylic acid, 5-phenyl-pyrrolidine-2-carboxylic acid, 3-pyridylalanine (3-Pya), 4-pyridylalanine (4-Pya), styrylalanine, tetrahydroisoquinoline-1-carboxylic acid (Tiq), 1 ,2,3,4-tetrahydronorharmane-3-carboxylic acid (Tpi), β-(2-thienry -alanine (Tha).
"Peptides" are selected from dipeptides to decapeptides, preferred are dipeptides, tripeptides, tetrapeptides and pentapeptides. The amino acids for the formation of the "peptides" can be selected from those listed above.
An "aza-amino acid" is defined as an amino acid where the chiral a-CH group is replaced by a nitrogen atom, whereas an "aza-peptide" is defined as a peptide, in which the chiral a-CH group of one or more amino acid residues in the peptide chain is replaced by a nitrogen atom.
Other amino acid substitutions for those encoded in the genetic code can also be included in peptide compounds within the scope of the invention and can be classified within this general scheme. Proteinogenic amino acids are defined as natural protein-derived oc-amino acids. Non-proteinogenic amino acids are defined as all other amino acids, which are not building blocks of common natural proteins.
"Peptide mimetics" per se are known to a person skilled in the art. They are preferably defined as compounds which have a secondary structure like a peptide and optionally further structural characteristics; their mode of action is largely similar or identical to the mode of action of the native peptide; however, their activity (e.g. as an antagonist or inhibitor) can be modified as compared with the native peptide, especially vis a vis receptors or enzymes. Moreover, they can imitate the effect of the native peptide (agonist). Examples of peptide mimetics are scaffold mimetics, non-peptidic mimetics, peptoides, peptide nucleic acids, oligopyrrolinones, vinylogpeptides and oligocarbamates. For the definitions of these peptide mimetics see Lexikon der Chemie, Spektrum Akademischer Verlag Heidelberg, Berlin, 1999.
The aim for using these mimetic structures is increasing the activity, increasing the selectivity to decrease side effects, protect the compound against enzymatic degradation for prolongation of the effect.
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 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 or solvate is also intended, provided such is possible or appropriate under the circumstances.
Salts and solvates of the compounds of formula (1) 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, sulphuric, nitric, citric, tartaric, phosphoric, lactic, pyruvic, acetic, trifluoroacetic, triphenylacetic, sulphamic, sulphanilic, succinic, oxalic, fumaric, maleic, malic, mandelic, glutamic, aspartic, oxaloacetic, methanesulphonic, ethanesulphonic, arylsulphonic (for example p-toluenesulphonic, benzenesulphonic, naphthalenesulphonic or naphthalenedisulphonic), 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.
Examples of solvates include hydrates.
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 and the patent applications DE 198 28 1 13, DE 198 28 14, WO 99/67228 and WO 99/67279 which are fully incorporated herein by reference.
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. cOmie, 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.
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.
Peptide Sequences The peptides mentioned and used herein have the following sequences: Αβ(1-42): Asp-Ala-Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-Val-His-His-Gln-Lys-Leu-Val-Phe-P e-Ala-Glu-Asp-Val-Gly-Ser-Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met-Val-Gly-Gly-Val-Val-lle-Ala Αβ(1-40): Asp-Ala-Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-Val-His-His-Gln-Lys-Leu-Val-Phe-P e-Ala-Glu-Asp-Val-Gly-Ser-Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met-Val-Gly-Gly-Val-Val Αβ(3-42): Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-Val-His-His-Gln-Lys-Leu-Val-Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser-Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met-Val-Gly-Gly-Val-Val-lle-AI^ Αβ(3-40): Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-Val-His-His-Gln-Lys-Leu-Val-Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser-Asn-Lys-Gly-Ala-!le-lle-Gly-Leu-Met-Val-Gly-Gly-Val-Val Ap(1-11)a: Asp-Ala-Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-NH2 A (3-11)a: Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-NH2 Αβ(1-21)3: Asp-Ala-Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-Val-His-His-Gln-Lys-Leu-Val-Phe-Phe-Ala-NH2 Ap(3-21)a: Glu-Phe-^Arg-His-Asp-Ser-Gly-Tyr-Glu-Val-His-His-Gln-Lys-Leu-Val-Phe-Phe-Ala-NH2 21 176765/2 ΘΙη3-Αβ(3-40): Gln-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-Val-His-Hts-Gln-Lys-Leu-Val-Phe-Ph Glu-Asp-Val-Gly-Ser-Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met-Val-Gly-Gly-Val-Val Gln3-AP(3-21)a: Gln-Phe-Arg-His-Asp-Ser-G!y-Tyr-Glu-Val-His-His-Gln-Lys-Leu-Val-Phe-Phe-AIa-NH2 Gln3-Ap(1-11)a: Asp-A!a-Gln-Phe-Arg-His-Asp-Ser-Giy-Tyr-Giu- Hz Gln3-AP(3-11)a: Gln-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-NH2 Summary of the invention The present invention provides compounds that act as inhibitors of glutaminyl cyclase (QC, EC 2.3.2.5).
Physiological substrates of QC in mammals are, e.g. [GIu3] amyloid β-protein (3-40/42), [Gin3] amyloid β-protein (3-40/42), Gastrin, Neurotensin, FPP, CCL 2, CCL 7, CCL 8, CCL 16, CCL 18, Fractalkine, Orexin A, [Gln3]-glucagon(3-29) and [Gln5]-substance P(5-1 ). The compounds according to the present invention and pharmaceutical compositions comprising at least one compound according to the present invention are useful in the manufacture of medicaments for the treatment of conditions that are to be treated by modulation of QC activity.
By administering inhibitors of QC (EC) activity to a mammal it is possible to prevent or alleviate or treat neuronal disorders (Alzheimer's disease, Down Syndrome, Parkinson disease, Chorea Huntington, pathogenic psychotic conditions, schizophrenia, impaired food intake, sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance, impaired regulation, body fluids, hypertension, fever, sleep dysregulation, anorexia, anxiety related disorders including depression, seizures including epilepsy, 22 176765/2 drug withdrawal and alcoholism, neurodegenerative disorders including cognitive dysfunction and dementia).
Furthermore, the medicaments containing the compounds of the present invention are capable to stimulate the proliferation of myeloid progenitor cells.
In addition, the medicaments containing QC inhibitor according to the present invention can lead to suppression of male fertility.
In a preferred embodiment, the present invention provides use of inhibitors of QC (EC) activity in combination with other agents, especially for the manufacture of medicaments for the treatment of neuronal disorders.
The present invention is directed to a compound of the formula 1 including pharmaceutically acceptable salts thereof, including all stereoisomers and polymorphs: N J--AA--BS formula 1 wherein: A is an unbranched C3 alkyl chain; and B is a group selected from (VI) or (VII): (VI) (VII) wherein: D represents substituted phenyl, wherein substitution means -oxyalkyl, -thioalkyl, halogenyl; or D represents dihydrobenzodioxine, benzodioxole, benzodithiole, dihydrobenzodithiine, benzooxathiole or dihydrobenzooxathiine; X represents S, N-CN; Y is S; 17 22a R17 and R18 are both H; one of R17 and R18 is H and the other is methyl; or R17 and R18 can be connected to form a carbocycle with up to 6 ring atoms; and wherein the term alkyl means methyl, ethyl, propyl or butyl, with the proviso (f) that the compounds 176765/1 are excluded from formula 1; or a compound of the formula la including pharmaceutically acceptable salts thereof, including all stereoisomers and polymorphs: wherein the group R represents methyl, teri-butyl, benzyl, phenyl, 4-ethyl-phenyl, 4-(methoxycarbonyl)-phenyl, 4-(methylcarbonyl)-phenyl, bicyclo[2.2.1]hept-5-en-2-yl, 2-(methoxycarbonyl)-phenyl, 4-(oxazol-5-yl)-phenyl, 4-(pyrazol-l-yl)-phenyl, 4-isopropyl-phenyl, 4-(piperidine-l-sulfonyl)-phenyl, 4-cyano-phenyl, 2,3-dihydro-benzo[l,4]-dioxin-6-yl, benzo[l,3]dioxol-5-yl, 4-(benzyloxy)-phenyl, 4-(methoxy)-benzyl, 4-methyl-phenyl, naphthalen-l-yl, 4-nitro-phenyl, n-butyl, cyclooctyl, furan-2-yl-methyl, tetrahydrofuran-2-yl-methyl, benzo[l,3]dioxol-5-ylmethyl, 2-(rra^holin-4-yl)-ethyl, 4-(dimethylamino)-phenyl, 4-(trifluoromethoxy)-phenyl, benzoyl or pyridin-4-yl; with the proviso (e) that the following compounds are excluded: or a compound of the formula lb including pharmaceutically acceptable salts thereof, including all stereoisomers and polyn ^hs: wherein the group R2 represents methyl, terf-butyl, benzyl, phenyl, 4-ethyl-phenyl, 4-(trifluoromethyl)-phenyl, 4-(methylcarbonyl)-phenyl, 2,3-dihydro-benzo[l ,4]-dioxin-6-yl, 22c 176765/1 benzo[d][l,3]dioxol-6-yl, 4-(ben2yloxy)-phenyl, 4-methyl-phenyl, naphthalen-l-yl, 4- nitro-phenyl, n-butyl, cyclooctyl, furan-2-yl-methyl, tetrahydrofuran-2-yl-methyl, benzo[d][l,3]dioxol-6-ylmethyl, 2-(mo holin-4-yl)-ethyl, 4-(dimethylamino)-phenyls or trityl.
The present invention is directed to a pharmaceutical composition comprising at least one compound according to the invnetion optionally in combination with a therapeutically acceptable carrier and/or excipient.
The pharmaceutical composition according to the invention may further comprise additionally at least one compound, selected from the group consisting of PEP-inhibitors, LiCl, inhibitors of dipeptidyl aminopeptidases, preferably inhibitors of DP IV or DP IV- like enzymes, NPY-receptor ligands, NPY agonists, ACE inhibitors, PHVIT enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of neutral endopeptidase, PDE-4 inhibitors, MAO inhibitors, TNFalpha inhibitors, amyloid protein or amyloid peptide deposition inhibitors, sigma-1 receptor inhibitors and histamine H3 antagonists.
Detailed Description of the Invention The present invention provides novel inhibitors of QC (EC) of the formula 1 , formula 1 wherein: A is either: an alkyl chain, alkenyl chain or alkynyl chain; a group selected from: (I) (II) (III) wherein: 23 R6, R7, R8, R9 and R10 are independently H or an alkyl chain, alkenyl chain, alkynyl chain, cycloalkyi, a carbocycle, aryl, heteroaryl, or a heterocycle; n and n1 are independently 1 - 5; m is 1 - 5; o is 0 - 4; and B is a group selected from (VI) - (XIV): (XIV) wherein: 24 D and E independently represent an alkyl chain, alkenyl chain, alkynyl chain, a cycloalkyl, carbocycle, aryl, -alkylaryl, heteroaryl, -alkylheteroaryl, acyl or a heterocycle.
X represents CR20R21, 0, S, NR19, with the proviso for formulas (VIII) and (IX) that, if Z = CH, X is O or S; R 9 is selected from the group consisting of H, alkyl, cycloalkyl, aryl, heteroaryl, -oxyalkyl, -oxyaryl, carbonyl, amido, hydroxy, N02) NH2, CN; R20 and R21 are independently selected from H, alkyl, cycloalkyl, heterocycle, aryl, heteroaryl, ,-oxyalkyl, -oxyaryl, carbonyl, amido, N02 , NH2, CN, CF3 ; X1, X2 and X3 are independently O or S provided that X2 and X3 are not both O; Y is O or S, with the proviso that Y may not be O, when the carbocycle formed by R 7 and R 8 has 3 members in the ring; Z is CH or N; R11, R 2, R13 and R 4 can be independently selected from H, an alkyl chain, an alkenyl chain, an alkynyl chain, cycloalkyl, carbocycle, aryl, heteroaryl, a heterocycle, halogen, alkoxy-, -thioalkyl, carboxyl, carboxylic acid ester, carbonyl, carbamide, carbimide, thiocarbamide or thiocarbonyl, NH2, N02; R15 and R16 are independently of each other H or a branched or unbranched alkyl chain, or a branched or unbranched alkenyl chain; R17 and R 8 are independently selected from H or an alkyl chain, alkenyl chain, a alkynyl chain, a carbocycle, aryl, heteroaryl , heteroalkyi or can be connected to form a carbocycle with up to 6 ring atoms; n is 0 or 1 ; with the proviso that the following compounds: and and and are excluded from formula 1 , When A is selected from an alkyl chain, alkenyl chain or alkynyl chain, preferably A is a C1-C7 alkyl chain, C1-C7 alkenyl chain or a C1-C7 alkynyl chain. In one embodiment of the invention A is an unbranched C2-5 alkyl chain, in particular an unbranched C3-alkyl chain, especially an unbranched C3 alkyl chain. In a second embodiment of the invention A represents a C3 alkyl chain which is substituted at the 2 position by one (i.e. in S or R configuration) or two methyl groups. 26 When A is selected from the formulae (I) to (V), preferably A is selected from groups (I) to (IV). In one embodiment of the invention A represents a group of formula (IV), wherein n1 are each equal to 1 and m = 1-4, especially m = 1. In a second embodiment of the invention A represents a group of formula (I), (ll> or (III), wherein n and n1 are each equal to 1 and R6, R7, RB, R9 and R10 represent H.
Preferably R6, R7, R8, R9 and R10 represent H or methyl.
In one embodiment of the invention the group B is chosen from (VI), (Via), (Vlb), (VII). (X). (Xl)> (Xll). (X'H) and (χιν)· ln a second embodiment of the invention group B represents formula (VI). In a third embodiment of the invention group B represents formula (Via). In a fourth embodiment of the invention group B represents formula (Vlb). In a fifth embodiment of the invention group B represents formula (VII). In a sixth embodiment of the invention group B represents formula When B represents a group (IX) suitably A does not represent alkynyl.
Preferably D and E independently represent benzyl, aryl, heteroaryl or a heterocycle.
In one embodiment of the invention D and E represent aryl, in particular phenyl or napthyl, especially substituted phenyl. Preferred substituent groups when D represents phenyl include alkoxy-, -thioalkyl, halogen, or a carbo:xylic acid alkyl or aryl ester. Also preferred are fluoro, chloro, bromo, iodo, trifluoromethyl, trifluoromethoxy, methoxy, ethoxy, benzyloxy, cyano, acetyl, dimethyl amino, methylsulphanyl, nitro, oxazolyl, pyrazolyl, isopropyl, ethyl and methoxycarbonyl. Where a phenyl group is mono-substituted it is preferred that substitution is in the 4-position. Other suitable aryl groups which D and E may represent include dihydrobenzodioxine, benzodioxole, benzodithiole dihydrobenzodithiine, 27 benzooxathiole and dihydrobenzooxathiine. A particularly preferred group which D or E may represent is 3,4- (dimethoxy)-phenyl, Preferably R20 and R21 represent N02, CN, CF3 or, if R20 is H, R2 is N02, CN, CF3l or, if R21 is H, R20 is N02, CN, CF3.
In one embodiment, X or Y is S, O or NR1. Preferably X or Y is S.
Preferably Z represents N.
In a preferred embodiment, R11 and R14are H.
In a further preferred embodiment, R12 and R13 are independently selected from oxyalkyi or thioalkyi, halogen, or carboxyiic acid alkyl ester or phenyl.
In a preferred embodiment, at least one of R15 and R16 is H, more preferably, R15 and R16 are both H.
In a preferred embodiment, one of R 7 and R 8 is H and the other is Me. Also preferred are compounds wherein one of R17 and R18 is H and the other is phenyl. Additionally preferred are compounds where R17 and R 8 form a carbocycle with up to 6 members in the ring atoms.
Preferred compounds include those defined by Examples 13, 119 and 125 below.
The present invention provides compounds of formula 1 for use as a pharmaceutical, with the proviso that the compounds: and and are excluded from formula 1.
The compound (a) of the proviso above is disclosed as compound 7 in Ganellin et al (1995) J Med Chem 38(17) 3342-3350. This paper discloses said compound as a weak inhibitor of the histamine H3 receptor.
The compound of proviso (b) is disclosed as compound 7 in Venkatachalam et al (2001) Bioorganic Med Chem Lett 11 , 523-528. This discloses said compound as an HIV1 reverse transcriptase inhibitor.
The compound of proviso (c) is disclosed as compound 19b in Moon et al (1991) J Med Chem 34, 2314-2327. This paper discloses said compound as a cholinergic agonist, with potential use in the treatment of Alzheimer's disease. 29 The compounds of proviso (d) are disclosed as compounds 99, 100 and 102-103 in Wright et al (1986) J Med Chem 29, 523-530. This paper discloses said compounds as thromoxane synthetase inhibitors.
Certain compounds which would be embraced by formula 1 if it were not for the proviso "provided that X2 and X3 are not both O" are disclosed in Wright et al (1987) J Med Chem 30, 2277-2283 as thromboxane synthetase inhibitors.
Certain compounds which would be embraced by formula 1 if it were not for the proviso "that Y may not be O, when the carbocycle formed by R17 and R18 has 3 members in the ring" are disclosed in EP 0 117 462 A2 as thromboxane synthetase inhibitors.
Furthermore, the present invention provides the use of inhibitors of QC of the formula 1 , without the proviso excluding compounds (a)-(d) or the proviso that X2 and X3 are not both O or the proviso that Y may not be O, when the carbocycle formed by R17 and R18 has 3 members in the ring, for the preparation of a medicament for the treatment of diseases selected from the group consisting of Alzheimer's disease, Down Syndrome, Parkinson disease, Chorea Huntington, pathogenic psychotic conditions, schizophrenia, impaired food intake, sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance, impaired regulation, body fluids, hypertension, fever, sleep dysregulation, anorexia, anxiety related disorders including depression, seizures including epilepsy, drug withdrawal and alcoholism, neurodegenerative disorders including cognitive dysfunction and dementia.
The present invention also provides inhibitors of QC of the formula 1 , without the proviso excluding compounds (a)-(d) or the proviso that X2 and X3 are not both O or the proviso that Y may not be O, when the carbocycle formed by R17 and R18 has 3 members in the ring, for use in the treatment of diseases selected from the group consisting of Alzheimer's disease, Down Syndrome, Parkinson disease, Chorea Huntington, pathogenic psychotic conditions, schizophrenia, impaired food intake, sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance, impaired regulation, body fluids, hypertension, fever, sleep dysregulation, anorexia, anxiety related disorders including depression, seizures including epilepsy, drug withdrawal and alcoholism, neurodegenerative disorders including cognitive dysfunction and dementia.
The present invention also provides a method of treatment for a disease selected from the group consisting of Alzheimer's disease, Down Syndrome, Parkinson disease, Chorea Huntington, pathogenic psychotic conditions, schizophrenia, impaired food intake, sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance, impaired regulation, body fluids, hypertension, fever, sleep dysregulation, anorexia, anxiety related disorders including depression, seizures including epilepsy, drug withdrawal and alcoholism, neurodegenerative disorders including cognitive dysfunction and dementia, comprising the administration of a therapeutically active amount of at least one compound of formula 1 , without the proviso excluding compounds (a)-(d) or the proviso that X2 and X3 are not both O or the proviso that Y may not be O, when the carbocycle formed by R17 and R18 has 3 members in the ring , to a mammal, preferably a human.
Most preferably, the present invention provides a method of treatment and corresponding uses for a disease selected from the group consisting of Alzheimer's disease, Down Syndrome, Parkinson disease and Chorea Huntington, comprising the administration of a therapeutically active amount of at least one compound of formula 1 , without the proviso excluding compounds (a)-(d), to a mammal, preferably a human.
Suitably in the above mentioned methods and uses the compound is not the compound of proviso (c).
A further compound, that of formula 1* shown below, is also a novel inhibitor of QC: 31 The compound of formula 1* may be employed in the methods and uses according to the invention in an analogous manner to the compounds of formula 1 described above.
In a further embodiment, the present invention provides novel inhibitors of QC (EC) of the formula 1a, wherein R is defined in examples 1 to 53.
ESI-MS Res.Act. ICso Ki Example R (M+H) (%) (μΜ) (μΜ) 1 Methyl 199.3 4.3 13 2 fert-Butyl 241.4 60.7 14.7 3 Benzyl 275.4 60.9 5.67 4 Phenyl 261.4 42.3 4.4 4-(fluoro)-phenyl 279.35 42.0 4.73 6 4-(chloro)-phenyl 295.80 1.2 7 4-(ethyl)-phenyl 289.41 28.7 2.78 8 4-(trifluoromethyl)-phenyl 329.4 38.5 3.93 4-(methoxy-carbonyl)- 319.4 9 1.19 Phenyl 4-(acetyl)-phenyl 303.4 17.0 1.70 11 4-(methoxy)-phenyl 291.4 9.7 0.70 32 ESI-MS ResAct. 1C50 K, Example R (M+H) (%) (μΜ) (MM) 12 bicyclo[2.2.1 ]hept-5-en-2-yl 277.5 16.0 13 3 ,4-(dimethoxy)-pheny I 321.5 0.7 0.22 0.06 14 2,4-(dimethoxy)-phenyl 321.5 2.2 0.57 3 , 5-(d imethoxy)-pheny I 321.5 2.86 0.75 2-(methoxy-carbonyl)- 319.4 16 Phenyl 17 4-(oxazol-5-y)-phenyl 328.5 3.64 0.86 18 4-(pyrazol-1 -yl)-phenyl 327.4 19 4-(isopropyl)-phenyl 303.5 8.7 4-(piperidine-1 -sulfonyl)- 408.6 8.5 2.27 Phenyl 21 4-(morpholin-4-yl)-phenyl 346.5 9.0 22 4-(cyano)-phenyl 286.4 9.0 2.89 2,3-dihydro-benzo[1 ,4] 319.4 4.17 23 1.12 dioxin-6-yl 24 benzo[1 ,3]dioxol-5-yl 305.4 16.7 5.66 3,4,5(trimethoxy)-phenyl 351.5 1.7 0.34 26 3-(methoxy)-phenyl 291.4 6.8 1.86 27 4-(ethoxy)-phenyl 305.5 7.2 0.89 28 4-(benzyloxy)-phenyl 367.5 0.98 29 4-(methoxy)-benzyl 305.5 3.93 3,4-(dimethoxy)-benzyl 335.5 1.55 2-(methoxy-carbonyl)- 325.5 31 thiophene-3-yl 3-(ethoxy-carbonyl)- 392.6 4,5,6,7- 32 tetrahydrobenzo[b]thio- phene2-yl 2-(methoxy-carbonyl)-4- 339.5 33 (methyl)-thiophene-3-yl 33 In a further embodiment, the present invention provides novel inhibitors of QC (EC) of the formula 1 b, 34 wherein R and R2 are defined in examples 54 to 95.
ESI-MS Res.Act. Ki Example R R2 (M+H) (%) (μΜ) 54 Cyano Methyl 207.3 1.5 55 Cyano 3,4-(dimethoxy)-phenyl 329.4 1.36 56 Cyano 2,4-(dimethoxy)-phenyl 329.4 57 Cyano 3 , 5-(d imethoxy)-phenyl 329.4 0.91 2,3- 58 Cyano dihydrobenzo[b][1 ,4]dioxin- 327.4 0.64 7-yl 59 Cyano Benzo[d][1 ,3]dioxol-6-yl 313.4 0.73 60 Cyano 3,4,5-(trimethoxy)-phenyl 359.4 0.88 61 Cyano 3-(methoxy)-phenyl 299.4 62 Cyano 4-(ethoxy)-phenyl 313.4 63 Cyano 4-(benzyloxy)-phenyl 375.5 64 Cyano Phenyl 269.4 1.02 65 Cyano 4-(methoxy)-phenyl 299.4 0.70 66 Cyano 4-(acetyl)-phenyl 311.4 67 Cyano 4-(nitro)-phenyl 314.4 68 Cyano Benzyl 283.4 22.5 8.17 69 Cyano Naphthalen-1-yl 319.4 70 Cyano 4-(fluoro)-phenyl 387.3 71 Cyano 4-(iodo)-phenyl 395.3 72 Cyano 4-(bromo)-phenyl 348.3 73 Cyano Cyclooctyl 289.4 74 Cyano ferf-butyl 249.3 75 Cyano 4-(methyl)-phenyl 283.3 1.34 In a further embodiment, the present invention provides novel inhibitors of QC (EC) of the formula 1 c, wherein R3 is defined in examples 96 to 102.
In a further embodiment, the present invention provides novel inhibitors of QC (EC) of the formula 1d, wherein the position on the ring is defined in examples 103 to 105.
In a further embodiment, the present invention provides novel inhibitors of QC (EC) of the formula 1e, 37 wherein R4 and R5 are defined in examples 106 to 109.
In a further embodiment, the present invention provides novel inhibitors of QC (EC) of the formula 1f, wherein R6 is defined in examples 110 to 112.
In a further embodiment, the present invention provides novel inhibitors of QC (EC) of the formula 1g, 38 wherein R7, R8and R9 are def ned in examples 113 to 132. 39 In a further embodiment, the present invention provides novel inhibitors of QC (EC) of the formula 1h, wherein n is defined in examples 133 to 135.
In a further embodiment, the present invention provides novel inhibitors of QC (EC) of the formula 1 i, wherein m is defined in examples 136 and 37 Further novel inhibitors of QC (EC) are examples 138 to 141.
ESI-MS Res.Act. IC50 κ, Example Structure (M+H) (%) (μΜ) (pM) 138 347.5 139 347.2 Synthesis of the examples Synthesis scheme 1: Synthesis of the examples 1-53, 96-102, 136-137 Reagents and conditions: (a) NaH, DMF, 4h, rt.; (b) , 8h, 100°C; (c) H2N-NH2lEtOH, 8h, reflux then 4N HCI, 6h, reflux, (d) R3-NCO, EtOH, 6h, reflux, (e) 3,4 dimethoxy-phenyl-isothiocyanate, Synthesis scheme 2: Synthesis of the examples 54-95 R1- H2 + 41 Reagents and conditions: (a) R-NCS, EtOH, 6h, reflux; (b) WSCD, 1H-imidazole-1-propanamine, DMF, 2h, r.t.
Synthesis scheme 3: Synthesis of the examples 103-105 Reagents and conditions: (a) NaH, DMF, rt.,3h; (b) UAIH4, dioxane, reflux, 1h; (c) R-NCS, EtOH, reflux 6h, Synthesis scheme 4: Synthesis of the examples 106 - Reagents and conditions: (a) EtOH, 2 h, reflux Synthesis scheme 5: Synthesis of the examples 110 - 112 Reagents and conditions: (a) 1H-imidazole-1-propanamine, Triethylamine, Toluene, 12 h, reflux 42 Synthesis scheme 6: Synthesis of the examples 113 -132 Reagents and conditions: (a) CAIBE, 1H-imidazole-1-propanamine, Dioxan, 0°C, 12h; (b) Laweson's Reaent, EtOH, reflux, 8h Synthesis scheme 7: Synthesis of the examples 133 - Reagents and conditions: (a) 1H-imidazole-1-propan acidic chloride, CH2CI2, - 10*0., 1 h; (b) Lawesson's Reagent, Dioxane, reflux, 8 h Synthesis scheme 8: Synthesis of the example 138 Reagents and conditions: (a) EtOH, reflux, 8 h Synthesis scheme 9: Synthesis of the example 139 Reagents and conditions: (a) 75% cone. H2SO4 , 4h 43 Synthesis scheme 10: Synthesis of the example 140 Reagents and conditions: (a) Acetonitrile, reflux 2h Synthesis scheme 11: Synthesis of the example 141 Reagents and conditions: (a) NaH, DMF, 4h, rt.; (b) , 8h, 100°C; (c) H2N-NH2lEtOH, 8h, reflux then 4N HCI, 6h, reflux, (d) 3,4 dimethoxy-phenyl- isothiocyanate, EtOH, 6h, reflux Analytical conditions ESI-Mass spectra were obtained with a SCIEX API 365 spectrometer (Perkin Elmer). The 1H-NMR (500 MHz) data was recorded on a BRUKER AC 500, using DMSO-D6 as solvent. Chemical shifts are expressed as parts per million downfield from tetramethylsilane. Splitting patterns have been designated as follows: s (singulet), d (doublet), dd (doublet of doublet), t (triplet), m (multiplet), and br (broad signal).
Detailed synthesis description Examples 1 - 12 and 14 - 53 1H-imidazole-1-propanamine was reacted with the corresponding isothiocyanate in ethanol under reflux for 8h. After that the solvent was removed and the remaining oil 44 was dissolved in methylene chloride. The organic layer was washed twice with a saturated solution of NaHC03 followed by NaHS04 and brine, dried then evaporated. The remaining solid was re-crystallized from ethyl acetate, yielding the example thiourea in yields of 80 - 98%.
.Example 13 1-(3-(1H-imidazol-1-yl)propyl)-3-(3,4-dimet oxyphen 4.0 mmol of 3,4-dimethoxyphenyl isothiocyanate and 4.0 mmol of 3-(1H-imidazol-1-yl)alkyl-1 -amine were dissolved in 10 mL of absolute ethanol. After stirring for 2 h under reflux, the solvent was evaporated and the resulting solid was recrystallized from ethanol.
Yield: 0.66 g (51.3 %); mp: 160.0 - 161.0°C 1H NMR 5 1.8 - 2.0 (m, 2H), 3.4 - 3.5 (m, 2H), 3.75 (s, 6H), 3.9 - 4.0 (m, 2H), 6.7 - 6.8 (m, 1H), 6.9 (br m, 2H), 6.95 (s, 1H), .7.15 (s, 1H), 7.55 (br s, 1H), 7.6 (s, 1H), 9.3 (s, 1 H); MS m/z 321.2 (M+H), 253.3 (M-C3H3N2') Examples 96 - 102 1H-imidazole-1-propanamine was reacted with the corresponding isocyanate in ethanol under reflux for 8h. After that the solvent was removed and the remaining oil was dissolved in methylene chloride. The organic layer was washed twice with a saturated solution of NaHC03 followed by NaHS04 and brine, dried then evaporated. The remaining solid was re-crystallized from ethyl acetate, yielding the example urea in yields of 85 - 90%.
Examples 136, 137 The 1H-imidazole-1-alkylamines were prepared according to the literature from ω-brom-alkyl-phtalimides and imidazolium salt and. subsequent hydrazinolysis. The resulting products were transformed into the thioureas according to example 1 - 53 giving a 88% (example 136) and 95% (example 137) yield.
Examples 54 - 95 All examples were made from the corresponding thioureas by reacting with Water-soluble-carbodiimide (VVSCD) and 1H-imidazole-1-propanamine in dry dimethyl form-amide for 2h at r.t. giving the trisubstituted guanidines with yields from 40 - 87%. 45 Examples 103 - 105 Imidazole was reacted with the corresponding brommethylphenylcyanide in DMF, utilizing 1 equivalent of NaH for 3h under rt, giving the 1H-imidazole-1-methylphenylcyanides. The solvent was removed and the resulting oil was re-dissolved in dioxane. The cyanides were converted in the corresponding amines using 1 equivalent of LiAIH . After adding a saturated solution of KHS04, dioxane was evaporated and the aqueous layer was extracted by means of CHCI3. The organic layer was concentrated in vacuo and the amine was converted in the corresponding thioureas according to example 1 - 53 giving a 78% (example 103) and 65% (example 104) and 81% (example 105) yield.
Examples 106 - 109 Starting from the corresponding methansulfonate-2-methylpropyl-phthalimides the amines were synthesized as described for the amines in example 13S - 137. The resulting products were transformed into the thioureas according to ex mple 1 - 53 giving example 106 - 109 in total yields of 25 - 30%.
Examples 110 - 112 1H-imidazole-1-propanamine was reacted with the corresponding 2-ch lorobenzo[d] thiazole in toluol for 24h at a temperature of 130°C. After removing the solvent and recristallization from methanol example 110 - 12 was yielded in an amount of 55 -65%.
Examples 113 - 118, 120 - 124 and 126 - 132 1H-imidazole-1-propanamine was reacted with the corresponding 2- henyl acetic acid in dry dioxane by adding one equivalent of CAIBE and N-methylmo rpholine at a temperature of 0°C. After 2h the mixture was allowed to warm to r.t. and the mixture was stirred for 12h. After removing the solvent the resulting oil was redissolved in methylene chloride and the organic layer was washed by means of an aqueous solution of NaHC03 and water, dried and the solvent was evaporated. The remaining oil was dissolved in dioxane adding Laweson's Reagent. After stirring for 12h a saturated solution of aHC03 was added. Dioxane was evaporated and the aqueous layer was extracted by means of ethyl acetate. The organic layer was separated, 46 dried and the solvent was evaporated. The remainig solid was crystallized from acetyl acetate/ether, giving 113 - 118, 120 - 124 and 126 - 132 with total yields of 62 -85%.
Example 119 N-(3-(1H-imidazol-1-yl)propyl)-2-(3,4 limethoxyphenyl)ethan A mixture of 4.0 mmol triethylamine and 4.0 mmol of 3-(1H-imidazol-1-yl)alkyl-1-amine 20 mL of dioxane was added drop wise to an ice cooled, stirred solution of 4.0 mmol of 2-(3,4-dimethoxyphenyl)acetyl chloride in 30 mL of dioxane. The mixture was allowed to warm to r.t., and then stirred for 1 h. After removing the solvent by reduced pressure, the residue was redissolved in 50 mL of dichloromethane. The organic layer was washed by means of 30 mL of saturated aqueous solution of NaHC03, and water. The organic solution was dried, filtered, and the solvent was removed under reduced pressure. After redissolving in 50 mL of dry dioxane 2.2 mmol of Lawesson's reagent was added, and the mixture was heated to 90°C and stirred for 8 h. The solvent was removed by reduced pressure, and the residue was redissolved in 50 mL of dichloromethane. The organic layer was washed three times by means of a saturated aqueous solution of NaHC03, followed three times by water, dried, filtered, and then the organic solvent was removed. The compound was purified by chromatography using a centrifugal-force-chromatography device, (Harrison Research Ltd.) utilizing silica plates of a layer thickness of 2 mm, and a CHCb/MeOH gradient as eluting system.
Yield: 0.14 g (10.6 %); melting point: 148.0 - 150.0°C 1H NMR δ 2.0 - 2.15 (br m, 2H), 3.4 - 3.5 (m, 2H), 3.7 (s, 6H), 6.75 - 6.8 (m, 2H), 4.1 -4.2 (m, 2H), 6.8 - 6.9 (m, 2H), 6.95 - 7.0 (m, 1H), 7.4 (s, 1H), 7.75 - 7.85 (br m, 1H), 8.6 (s, 1H), 10.2 (s, 1H); MS m/z 320.2 (M+H), 252.2 (M-C3H3N2-) Example 125 N-(3-(1H-imidazol-1-yl)pmpyl)-1-(3,4-dimethoxyphenyl)cyclopropanec 11.06 mmol of 3,4-dimethoxyphenyl acetonitrile, 34.8 mmol of 2-Bromo-1-chloroethanole and 1.16 mmol of triethylbenzylammonium hydrochloride were dissolved in 10 mL of an aqueous soluton of KOH (60%). The mixture was transferred into an ultrasonic bath and vigorously stirred for 3h at room temperature. 47 The resulting suspension was diluted with 40 mL of water and extracted three times by means of 20 mL of dichloromethane. The combined organic layers where washed by means of an aqueous solution of hydrochloric acid (1 ), dried over Na2S04 and the solvent was removed under reduced pressure. The remaining oil was purified by flash-chromatography using silica gel and ethyl acetate/heptane as eluting system, resulting in 0.81 g (34.4 %) of 1-(3,4-dimethoxyphenyl)cyclopropanecarbonitrile 3.9 mmol of 1-(3,4-dimethoxyphenyl)cyclopropanecarbonitrile and 11.2 mmol of KOH were suspended in 80 mL of ethylene glycol. The mixture was stirred for 12 h under reflux. Then 80 mL of water were added and the aqueous layer was extracted two times with ether. After pH adjustment to a value of pH = 4 - 5 using HCI (1 N) the aqueous layer was extracted three times by means of ether, then the combined organic layers were dried over Na2S0 and the solvent was removed, resulting in 0.81 g (93.5%) of 1-(3,4-dimethoxyphenyl)cyclopropanecarboxylic acid. 3.44 mmol of 1-(3,4-dimethoxyphenyl)cyclopropanecarboxylic acid, 3.5 mmol of N-Methyl morpholine, and 3.5 mmol of isobutyl chloroformiat were dissolved in dry tetrahydrofurane and stirred for 5 min at -15°C. Then 3.5 mmol of 3-(1H-imidazol-1-yl)alkyl-1 -amine was added and the mixture was allowed to warm to 0°C and was stirred for 12h. The solvent was removed under reduced pressure and the remaining oil was redissolved in chloroform. Then the organic layer was washed two times by means of a saturated aqueous solution of NaHC03, then dried over a2S04 and the solvent was removed. Purification was performed by means of centrifugal forced chromatography using a chromatotron® device (Harrison Research Ltd.) utilizing silica plates of a layer thickness of 2 mm, and a CHCVMeOH gradient as eluting system resulting in 0.671 g (59.3%) of N-(3-(1 H-imidazol-1-yl)propyl)-1-(3,4-dimethoxyphenyl)cyclopropane-carboxamide.
After redissolving in 30 mL of dry dioxane 1.43 mmol of Lawesson's reagent were added, and the mixture was heated to 90°C and stirred for 8h. The solvent was removed by reduced pressure, and the residue was remains were dissolved in 50 mL of dichloromethane. The organic layer was washed three times by means of a saturated aqueous solution of NaHC03, followed three times by water, dried, filtered, and then the organic solvent was removed. The compound was purified by chromatography using a centrifugal-force-chromatography device, (Harrison 48 Research Ltd.) utilizing silica plates of a layer thickness of 2 mm, and a CHCb/MeOH gradient as eluting system.
Yield: 0.33 g (46.2 %); melting point: 127.0 - 127.5°C 1H NMR δ 1.1 - 1.2 (t, 2H), 1.55 - 1.6 (t, 2H), 2.0 - 2.1 (m, 2H), 3.5 - 3.6 (m, 2H), 3.7 -3.8 (s, 6H), 4.1 - 4.2 (t, 2H), 6.8 - 6.9 (m, 3H), 7.65 (s, 1H), 7.75 (s, 1H), 8.8 (m, 1H), 9.05 (s, 1 H; MS m/z 346.0 (M+H), 278.2 (M-C3H3N2'), 77.1 (M-C6H8N3S«) Examples 133 - 135 A mixture of 1 equivalent triethylamine and 3,4-dimethoxyaniline in dioxane was added to an stirred solution of the corresponding ω-bromoalkyl acidic chloride at a temperature of 0°C. The solution was allowed to warm to r.t. and stirred for 2h. The solvent was evaporated, and the remaining oil was redissolved in dichloromethane. The organic layer was washed by means of water, dried, filtered, and the solvent was removed under reduced pressure.
Imidazole and sodium hydride were suspended in and the mixture was stirred under inert conditions at r.t. for 3 h. ro-Bromo-N-(3,4-dimethoxy-phenyl)alkylamide was added and the mixture was heated to 100°C and stirred for 8 h. After that, the solvent was evaporated, hot toluene were added and the solution was filtered. Then the solvent was removed under reduced pressure. The transformation into the thioamides was performed as described for example 113 - 132 by means of Laweson's reagent, giving 133 - 135 in total yields of 13 - 20 %.
The analytical data for further examples, which were syntesized according to the general synthesis schemes described above, are as follows: Example 1: 1-(3-(1H-imidazol-1-yl)propyl)-3-methylthiourea melting point: 122 - 122.5°C H NMR δ 1.85 - 1.95 (m, 2H), 2.8 (s, 3H), 3.2 - 3.5 (br d, 2H), 3.8 - 3.9 (m, 2H), 6.85 (d, 1H), 7.15 (d, 1H), 7.3 - 7.5 (br d, 2H), 7.65 (s, 1 H); MS m/z 199.1 (M+H), 221.3 (M+Na), 131.0 (M-C3H3N2») Example 2: 1-(3-(1H-imidazol-1-yl)propyl)-3-tert-butylthiourea melting point: 147.0 - 147.5°C 49 H NMR δ 1.3 - 1.4 (s, 9H), 1.85 -1.95 (m, 2H), 3.5 (t, 2H), 3.8 (t, 2H), 6.85 (d, 1 H), 7.15 (d, 1H), 7.3 - 7.5 (br d, 2H), 7.65 (s, 1H); MS m/z 241.1 (M+H), 173.1 (M-C3H3N2») Example 3: 1-(3-(1H-imidazol-1-yl)propyl)-3-benzylthiourea melting point: 127.0 - 128.0°C 1H NMR δ 1.85 - 1.95 (m, 2H), 3.2 - 3.5 (br d, 2H), 3.8 - 3.9 (m, 2H), 4.6 (s, 2H), 6.8 (d, 1 H), 7.15 (d, 1H), 7.19 - 7.35 (m, 5H), 7.5 - 7.6 (br d, 2H), 7.85 (s, 1H); MS m/z 275.3 (M+H), 207.1 (M-C3H3N2») Example 5: 1-(3-(1H-imidazol-1-yl)propyl)-3-phenylthiourea melting point: 166.5 - 167.0°C 1H NMR δ 1.95 - 2.05 (m, 2H), 3.3 - 3.5 (br d, 2H), 3.9 - 4.0 (m, 2H), 6.85 (d, 1H), 7.05 (m, 1 H) 7.15 (d, 1H), 7.25 (m, 2H), 7.35 (m, 2H), 7.6 (s, 1 H), 7.8 (br s, 1H), 9.5 (br s, 1 H); MS m/z 261.1 (M+H), 193.2 (M-C3H3N2») Example 6: 1-(3-(1H-imidazol-1-yl)pivpyl)-3-(4-fluomphenyl)thiourea melting point: 147.0 - 148.0°C 1H NMR δ 1.95 - 2.05 (m, 2H), 3.3 - 3.5 (br d, 2H), 3.9 - 4.05 (m, 2H), 6.85 (d, 1 H), 7.05 - 7.15 (m, 3H), 7.3 - 7.4 (m, 2H), 7.6 (s, 1H), 7.7 - 7.8 (br s, 1H), 9.4 (br s, 1H); MS m/z 279.3 (M+H), 211.2 (M-C3H3N2«) Example 7: 1-(3-(1H-imidazol-1-yl)pmpyl)-3-(4-ethylphenyl)thiourea melting point: 100.0 - 100.5°C 1H NMR δ 1.15 - 1.2 (t, 3H), 1.9 - 2.0 (m, 2H), 2.5 - 2.6 (m, 2H), 3.3 - 3.5 (br d, 2H), 3.9 - 4.05 (m, 2H), 6.85 (d, 1 H), 7.1 - 7.2 (m, 3H), 7.25 - 7.3 (m, 2H), 7.6 (s, 1 H), 7.7 -7.8 (br s, 1H), 9.4 (br s, 1H); MS m/z 289.3 (M+H), 221.1 (M-C3H3N2») Example 8: 1-(3-(1 H-imidazol-1-yl)propyl)-3-(4-(tn^uoromethyl)phenyl)thiourea melting point: 154.5 - 155.0°C 1H NMR δ 1.9 - 2.1 (br m, 2H), 3.4 - 3.6 (br d, 2H), 3.95 - 4.1 (br m, 2H), 6.85 (d, 1 H), 7.2 (d, 1H), 7.6 - 7.8 (m, 5H), 8.2 (br s, 1H), 9.9 (br s, 1H); MS m/z 329.3 (M+H), 261.2 (Μ-03Η3Ν2·) 50 Example 10: 1-(3-(1H-imidazol-1-y prOpyl)-3-(4-acetylpheny[)thiourea melting point: 170.0 - 171.0°C 1H NMR δ 1.9 - 2.1 (br m, 2H), 2.4 - 2.5 (s, 3H), 3.2 - 3.5 (br m, 2H), 3.9 - 4.1 (m, 2H), 6.85 (d, 1H), 7.15 (d, 1H), 7.5 - 7.65 (br m, 3H), 7.8 - 7.9 (m, 2H), 8.1 (m, 2H), 9.8 (br s, 1 H); MS m/z 303.2 (M+H), 235.1 (M-C3H3N2«) Example 11 : 1-(3-(1H-imidazol-1-yl)propyl)-3-(4-methoxyphenyl)thiourea melting point: 125.0 - 125.5°C 1H NMR δ 1.8 - 2.0 (br m, 2H), 3.2 - 3.5 (br m, 2H), 3.7 (s, 3H), 3.9 - 4.0 (m, 2H), 6.7 -6.9 (m, 3H), 7.1 - 7.2 (m, 3H), 7.5 (s, 1H), 7.6 (s, 1H), 9.2 (s, 1H); MS m/z 291.1 (M+H), 223.2 (M-C3H3N2») Example 14: 1-(3-(1H-imidazol-1-yl)pmpyl)-3-(2,4-dimethoxyphenyl)thfo^ melting point: 120.0 - 120.5°C H NMR δ 1.8 - 2.0 (br m, 2H), 3.4 - 3.5 (br m, 2H), 3.75 (s, 6H), 3.9 - 4.0 (m, 2H), 6.5 (d, 1H), 6.6 (s, 1H), 6.9 (s, 1H), 7.15 (s, 1H), 7.3 (d, 1H), 7.5 (br s, 1H), 7.6 (s, 1H), 9.75 (s, 1H); MS m/z 321.2 (M+H), 253.3 (M-C3H3N2») Example 15: 1-(3-(1H'imidazol-1-yl)propyl)-3-(3,5-dimethoxyphenyl)thiourea melting point: 142.0 - 143.0°C H NMR δ 1.8 - 2.0 (br m, 2H), 3.4 - 3.5 (br m, 2H), 3.6 (s, 6H), 3.95 - 4.0 (m, 2H), 6.25 (m, 1 H), 6.6 (m, 2H), 6.9 (s, 1H), 7.2 (s, 1H), 7.6 (s, 1H), 7.8 (s, 1H), 9.5 (s, 1H); MS m/z 321.2 (M+H), 253.3 (M-C3H3N2») Example 23: 1-(3-(1H-imidazol-1-yl)propyl)-3-(2,3-dihydrobenzoM1,^^ thiourea melting point: 103.0 - 103.5°C 1H NMR δ 1.9 - 2.0 (br m, 2H), 3.3 - 3.5 (br d, 2H), 3.9 - 4.0 (m, 2H), 4.2 - 4.3 (m, 4H), 6.7 (m, 1 H), 6.8 - 6.8 (m, 1 H), 6.9 (m, 2H), 7.2 (s, 1H), 7.6 (m, 2H), 9.3 (s, 1H); MS m/z 319.3 (M+H), 251.3 (M-C3H3N2») Example 24: 1-(3-( 1 H-imidazol- 1 -yl)propyl)-3-(benzo[d][ 1, 3Jdioxol-6-yl) thiourea melting point: 115.0 - 115.6°C 51 1H NMR δ 1.9 - 2.1 (br m, 2H), 3.4 - 3.5 (br d, 2H), 4.05 - 4.15 (m, 2H), 6.0 (s, 2H), 6.7 (m, 1H), 6.8 - 6.85 (m, 1H), 6.95 (d, 1 H), 7.25 (s, 1H), 7.45 (s, H), 7.7 (br s, 1H), 8.5 (br s, 1H), 9.4 (br s, 1H); MS m/z 305.2 (M+H), 237.2 (M-C3H3N2-) Example 25: 1-(3-(1H-imidazol-1'yl)pmpyl)-3-(3 5-trimethoxyphenyl)thioum melting point: 24.5 - 125.5°C 1H NMR δ 1.8 - 2.0 (m, 2H), 3.4 - 3.5 (br m, 2H), 3.6 (s, 3H), 3.7 (s, 6H), 3.9 - 4.0 (m, 2H), 6.65 (m, 2H), 6.85 (s, 1H), 7.2 (s, 1 H), 7.6 (s, 1H), 7.7 (br s, 1 H), 9.4 (s, 1H); MS m/z 351.3 (M+H), 283.2 (M-C3H3N2») Example 26: 1-(3-(1H-imidazol-1-yl)propyl)-3-(3-methoxyphenyl)thiourea melting point: 89.5 - 90.0°C 1H NMR δ 1.9 - 2.1 (br m, 2H), 3.4 - 3.5 (br m, 2H), 3.7 (s, 3H), 3.9 - 4.0 (m, 2H), 6.6 -6.7 (m, 1 H), 6.8 - 6.9 (m, 2H), 7.1 (m, 2H), 7.15 - 7.25 (br m, 1H), 7.6 (s, 1 H), 7.8 (br s, 1 H), 9.5 (s, 1 H); MS m/z 291.1 (M+H), 223.2 (M-C3H3N2«) Example 27: 1 -(3-(1H-imidazol-1-yl)propyl)-3-(4-ethoxyphenyl)thiourBa melting point: 126.0 - 126.5°C 1H NMR δ 1.5 (br m, 3H), 1.9 - 2.0 (br m, 2H), 3.4 - 3.5 (br m, 2H), 3.9 - 4.0 (br m, 4H), 6.8 - 6.9 (m, 2H), 6.95 (s, 1 H), 7.15 - 7.2 (m, 2H), 7.25 (s, 1 H), 7.55 - 7.6 (br s, 1 H), 7.8 (s, 1 H), 9.3 (s, 1 H); MS m/z 305.2 (M+H), 237.2 (Μ-03Η3Ν2·) Example 23 1-(3-(1H-imidazol-1-yl)pmpyl)-3-(4-(methylthio)phen melting point: 140.0 - 140.5°C 1H NMR δ 1.8 - 2.05 (br m, 2H), 2.5 (s, 3H), 3.3 - 3.5 (br m, 2H), 3.9 - 4.1 (m, 2H), 6.9 (m, 1H), 7.1 - 7.3 (br m, 5H), 7.6 (s, 1H), 7.75 (br s, 1H), 9.4 (s, 1 H); MS m/z 307.2 (M+H), 239.2 (M-C3H3N2«) Example 42: 1-(3-(1H-imidazol-1-yl)propyl)-3'(4-nitiOphenyl)thiourea melting point: 165.0 . 166.0°C 1H NMR δ 1.9 - 2.05 (m, 2H), 3.3 - 3.5 (br d, 2H), 3.95 - 4.05 (m, 2H), 6.85 (d, 1H), 7.15 (d, 1 H), 7.6 (d, 1H), 7.7 (m, 2H), 8.1 (m, 2H), 8.3 (br s, 1H), 10.1 (br s, 1H); MS m/z 306.2 (M+H), 237.9 (M-C3H3N2») 52 Example 50: 1-(3-(1H-imidazol-1-yl)pmpyl)-3-(4-(dimethylamino)phen melting point: 146.5 - 147.0°C 1H NMR δ 1.9 - 2.0 (m, 2H), 2.9 (s, 6H), 3.4 (m, 2H), 3.9 - 4.0 (m, 2H), 6.7 (m, 2H), 6.9 (s, 1H), 7.05 - 7.1 (m, 2H), 7.15 (s, 1H), 7.4 (br s, 1H), 7.6 (s, 1H), 9.2 (s, 1H); MS m/z 304.2 ( +H), 236.0 (M-C3H3N2') Example 102: 1-(3-(1H-imidazol-1-yl)pmpyl)-3-(3,4-dimethoxyphenyl)urea melting point: 114.5 - 115.0°C 1H NMR δ 1.7 - 1.9 (m, 2H), 2.9 - 3.1 (m, 2H), 3.7 (2s, 6H), 3.9 - 4.0 (m, 2H), 6.1 (t, 1 H), 6.7 (s, 2H), 6.8 (s, 1 H), 7.15 (d, 2H), 7.6 (s, 1H), 8.2 (s, 1H); MS m/z 321.2 (M+H), 253.3 (M-C3H3N2«) Example 106: 1-((S)-3-(1H-imidazol- 1-yl)-2-methylpropyl)-3-(3, 4-dimethoxyphenyl)-thiourea melting point:: 150.5 - 151.5°C 1H NMR δ 0.9 (d, 3H), 2.3 - 2.4 (m, 2Η), 2.5 (s, 1Η), 3.7 (d, 6H), 4.0 - 4.1 (br m, 1H), 4.15 - 4.25 (br m, 1H), 6.75 - 6.8 (m, 1H), 6.85 (m, 1 H), 6.9 - 7.0 (m, 1H), 7.65 (s, 1 H), 7.75 (s, 2H), 9.1 (s,1H), 9.5 (s, 1H); MS m/z 335.6 (M+H), 267.1 (M-C3H3N2-) Example 107: 1-((R)-3-(1H-imidazol-1-yl)-2-methylp pyl)-3-(3,4-dim^ thiourea melting point: 155.0 - 157.5 °C 1H NMR δ 0.9 (d, 3H), 2.3 - 2.4 (m, 2H), 2.5 (s, 1H), 3.7 (d, 6H), 4.0 - 4.1 (br m, 1H), 4.15 - 4.25 (br m, 1 H), 6.75 - 6.8 (m, 1 H), 6.85 (m, 1 H), 6.9 - 7.0 (m, 1H), 7.65 (s, 1 H), 7.75 (s, 2H), 9.1 (s,1 H), 9.5 (s, 1H); MS m/z 335.4 (M+H), 267.2 (M-C3H3N2») Example 109: 1-((1-((1H-imidazol- 1-yl)methyl)cyclopropyl)methyl)-3-(3, 4-dimethoxy-phenyl)thiourea melting point: 166.5 - 168.5°C 1H NMR δ 0.7 - 0.8 (br m, 2H), 1.85 - 1.9 (m, 1H), 2.15 - 2.2 (m, 1H), 2.2 - 2.3 (m, 1H), 3.4 - 3.5 (m, 1H), 3.7 (d, 6H), 4.2 (s, 1 H), 4.95 (s, 1H), 6.75 - 6.8 (br m, 1H), 6.85 - 6.9 (br m, 1H), 7.0 (s, 1H), 7.5 (m, 1H), 7.6 (m, 1H), 7.7 (s, 0.5H), 7.8 (s, 0.5H), 8.85 (s, 0.5 H), 9.1 (s, 0.5H), 9.35 (s, 0.5H), 9.45 (s, 0.5H); MS m/z 347.2 (M+H), 279.2 (M-C3H3N2'), 137.5 (M-C9H13N4S») 53 Example 110: N-(3-(1 H-imidazol- 1-yl) ropyl) benzo[d]thiazol-2-amine 1H NMR δ 1.95 - 2.15 (m, 2H), 3.25 - 3. 35 (m, 2H), 4.0 - 4.1 (t, 2H), 6.9 (s, 1H), 6.95 - 7.05 (t, 1H), 7.15 - 7.2 (m, 2H), 7.35 - 7.4 (d, 1H), 7.60 - 7.70 (m, 2H), 8.0 - 8.1 (br s, 1 H); MS m/z 259.4 (M+H), 191.3 (M-C3H3N2*) Example 111: N-(3-(1H-imidazol-1-yl)pmpy -6-chlorobenzo[d]thiazol-2-amine 1H NMR δ 1.95 - 2.15 (m, 2H), 3.25 - 3. 35 (m, 2H), 4.0 - 4.1 (t, 2H), 6.9 (s, 1 H), 7.1 -7.2 (d, 2H), 7.3 - 7.4 (d, 1 H), 7.65 (s, 1H), 7.8 (s, 1H), 8.2 (s, 1H); MS m/z 293.3 (M+H), 225.3 (M-C3H3N2«) Example 112: N-(3-(1H-imidazol-1-yl)propyl)-6-methoxybenzo[d]thiazol-2-am ^ H NMR δ 1.9 - 2.05 (m, 2H), 3.2 - 3. 3 (m, 2H), 3.7 (s, 3H), 4.0 - 4.1 (t, 2H), 6.7 - 6.8 (d, 1 H), 6.9 (s, 1H), 7.15 - 7.2 (s, 1 H), 7.2 - 7.3 (m, 2H), 7.65 (s, 1H), 7.8 (s, 1 H); MS m/z 289.1 (M+H), 221.4 (M-C3H3N2») Example 115: (R)Ή-(3-(1H-imidazol-1-yl)pmpyl)-2φhenyl m anethioamide melting point: 82.0 - 82.5°C H NMR δ 1.4 - 1.55 (d, 3H), 1.9 - 2.0 (m, 2H), 3.4 - 3.5 (m, 2H), 3.85 - 3.95 (m, 2H), 4.0 - 4.1 (q, 1H), 6.8 - 6.9 (s, 1H), 7.1 (s, 1H), 7.15 - 7.2 (m, 1H), 7.2 - 7.3 (m, 2H), 7.35 - 7.4 (m, 2H), 7.55 (s, 1H), 10.1 (s, 1H); MS m/z 274.4 (M+H), 206.3 (M-C3H3N2») Example 116: (S)-N-(3-(1H-i idazol-1-yI)pmpyl)-2^henylpmpanethioamide melting point: 82.5 - 83.5°C 1H NMR δ 1.4 - 1.55 (d, 3H), 1.9 - 2.0 (m, 2H), 3.4 - 3.5 (m, 2H), 3.85 - 3.95 (m, 2H), 4.0 - 4.1 (q, 1H), 6.8 - 6.9 (s, 1H), 7.1 (s, 1H), 7.15 - 7.2 (m, 1H), 7.2 - 7.3 (m, 2H), 7.35 - 7.4 (m, 2H), 7.55 (s, 1 H), 10.1 (s, 1 H); MS m/z 274.4 (M+H), 206.3 (M-C3H3N2-) Example 121 : N-(3-(1H-imidazol-1-yl)propyl)-1-(4-chlorophenyl)cyclobutanecarbO' thioamide melting point: 137.5 - 139.0°C 54 1H NMR δ 1.55 - 1.75 (br m, 2H), 1.85 - 1.95 (br m, 2H), 2.4 - 2.5 (br m, 2H), 2.7 -2.85 (br m, 2H), 3.3 - 3.5 (br m, 2H), 3.8 (m, 2H), 6.9 (s, 1H), 7.0 (s, 1H), 7.3 (m, 2H), 7.45 (s, 1H), 7.5 (m, 2H), 9.6 (t, 1 H); MS m/z 334.3 (M+H), 266.1 (M-C3H3N2») Example 122: N-(3-(1H-im}dazo -yl)propyl)-1-(4-chlomphenyl)cyclopentanecaώo-thioamide melting point: 140.0 - 141.0°C H NMR δ 1.5 - 1.65 (br m, 4H), 1.8 - 1.9 ( m, 2H), 2.0 - 2.1 (m, 2H), 2.6 (m, 2H), 3.4 -3.5 (m, 2H), 3.7 - 3.8 (m, 2H), 6.85 (s, 1 H), 7.0 (s, 1H), 7.35 (m, 2H), 7.4 (m, 2H), 7.5 (s, H), 9.4 (t, 1 H); MS m/z 348.2 (M+H), 280.2 (M-C3H3N2») Example 123: N-(3-(1H-imidazol-1-yl)propyl)-1-(4-methoxyphenyl)cyclohexanecarbo-thioamide melting point: 62.5 - 164.0°C 1H NMR δ 1.2 - 1.3 (m, 1 H), 1.35 - 1.5 (br m, 5H), 1.85 - 2.0 (br m, 4H), 2.4 - 2.6 (br m, 2H), 3.4 - 3.5 (m, 2H), 3.7 (s, 3H), 3.8 (m, 2H), 6.8 (m, 3H), 7.0 (s, 1H), 7.3 (m, 2H), 7.5 (s, 1H), 9.2 (t, 1 H); MS m/z 358.3 (M+H), 290.3 (M-C3H3N2«) Example 124: N-(3-(1 H-imidazol- 1-yl)pnopyl)- 1-(4-methoxyphenyl)cyclopropanecar-bothioamide melting point:: 129.0 - 129.5°C H NMR δ 1.0 - 1.1 (m, 2H), 1.5 - 1.6 (m, 2H), 1.9 - 2.0 (br m, 2H), 3.4 - 3.5 (m, 2H), 3.7 (s, 3H), 3.9 (m, 2H), 6.9 (m, 3H), 7.1 (s, 1H), 7.2 - 7.3 (m, 2H), 7.6 (s, 1H), 8.9 (br s, 1H); MS m/z 316.0 (M+H), 248.4 (M-C3H3N2-) Example 134: 5-( 1 H-imidazol-1-yl)-N-(3, 4-dimethoxyphenyl)pentanethioamide melting point:: 128.0 - 128.5°C 1H NMR δ 1.65 - 1.70 (m, 2H), 1.75 - 1.80 (m, 2H), 2.7 - 2.75 (m, 2H), 3.7 (s, 3H), 3.75 (s, 3H), 4.0 - 4.05 (t, 2H), 6.9 - 7.0 (m, 2H), 7.2 (s, 1H), 7.3 (d, 1H), 7.5 (s, 1H), 7.75 (s, 1 H), 11.0 (s, 1 H); MS m/z 320.2 (M+H), 252.2 (M-C3H3N2-) Example 136: 1-(2-(1H-fmidazol-1-yl)ethyl)-3-(3, 4-dimethoxyphenyl)thiourea melting point: 157.5 - 159.0°C 55 H N R δ 3.7 (2 s, 6H), 3.8 (m, 2H), 4.2 (m, 2H), 6.7 (m, 1H), 6.85 (m, 1H), 6.9 (m, 2H), 7.15 (s, 1H), 7.5 (br s, 1H), 7.6 (s, 1H), 9.5 (s, 1H); MS m/z 307.2 (M+H), 239.1 (M-C3H3N2») Example 137:1 -(4-(1 H-imidazol-1 -yl)butyl)-3-(3,4-dimethoxyphenyl)thiourea 45 melting point: 114.5 - 116.0°C 1H NMR δ 1.4 - 1.5 (m, 2H), 1.6 - 1.7 (m, 2H), 3.4 - 3.5 (m, 2H), 3.6 - 3.8 (br s, 6H), 3.9 - 4.0 (m, 2H), 6.7 (m, 1H), 6.9 (m, 2H), 6.95 (s, 1H), 7.2 (s, H), 7.6 (br s, 1H), 7.7 (s, 1H), 9.3 (s, 1H); MS m/z 335.3 (M+H), 267.1 (M-C3H3N2«) Physiological substrates of QC (EC) in mammals are, e.g. [Glu3] amyloid β-protein (3-40/42), [Gin3] amyloid β-protein (3-40/42), Gastrin, Neurotensin, FPP, CCL 2, CCL 7, CCL 8, CCL 16, CCL 18, Fractalkine, Orexin A, [Gln3]-glucagon(3-29) and [Gln5]-substance P(5-11). 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 is known to be cyclized to final pGlu Peptide Amino acid sequence Function Gastrin 17 QGPWL EEEEEAYGWM DF Gastrin stimulates the (amide) stomach mucosa to produce Swiss-Prot: P01350 and secrete 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. 56 Peptide Amino acid sequence Function Neurotensin QLYENKPRRP YIL Neurotensin plays an endocrine or paracrine role Swiss-Prot: P30990 in the 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 Swiss-Prot: P20396 the anterior pituitary gland and as a neurotransmitter/ neuromodulator in the central and peripheral nervous systems.
GnRH QHWSYGL P(G) amide Stimulates the secretion of gonadotropins; it stimulates Swiss-Prot: P01148 the secretion of both luteinizing and follicle- stimulating hormones. 57 Peptide Amino acid sequence Function CCL16 (small QPKVPEW VNTPSTCCLK Shows chemotactic activity inducible cytokine YYEKVLPRRL WGYRKALNC for lymphocytes and A16) HLPAIIFVTK RNREVCTNPN monocytes but not DDWVQEYIKD PNLPLLPTRN neutrophils. Also shows Swiss-Prot: LSTVKIITAK NGQPQLLNSQ potent myelosuppressive 015467 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 inducible cytokine RKIPIQRLES YTRITNIQCP attracts monocytes, A8) KEAVIFKTKR GKEVCADPKE lymphocytes, basophils and RWVRDSMKHL DQIFQNLKP eosinophils. May play a role Swiss-Prot: P80075 in neoplasia and inflammatory host responses. This protein can bind heparin. 58 Peptide Amino acid sequence Function CCL2 (small QPDAINA PVTCCYNFTN Chemotactic factor that inducible cytokine RKISVQRLAS YRRITSSKCP attracts monocytes and A2) KEAVIFKTIV AKEICADPKQ basophils but not neutrophils KWVQDSMDHL DKQTQTPKT or eosinophils. Augments Swiss-Prot: P 13500 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 inducible cytokine QKFIVDYSET SPQCPKPGVI attracts lymphocytes but not A18) LLTKRGRQIC ADPNKKWVQK monocytes or granulocytes.
YISDLKLNA May be involved in B cell Swiss-Prot: P55774 migration into 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. 59 Peptide Amino acid sequence Function Fractalkine QHHGVT KCNITCSKMT The soluble form is (neurotactin) SKIPVALLIH YQQNQASCGK chemotactic for T cells and RAIILETRQH RLFCADPKEQ monocytes, but not for Swiss-Prot: P78423 WVKDAMQHLD RQAAALTRNG neutrophils. The membrane- GTFEKQIGEV KPRTTPAAGG bound form promotes MDESWLEPE ATGESSSLEP adhesion of those leukocytes TPSSQEAQRA LGTSPELPTG to endothelial cells. May play VTGSSGTRLP PTPKAQDGGP a role in regulating leukocyte VGTELFRVPP VSTAATWQSS adhesion and migration APHQPGPSLW AEAKTSEAPS processes at the TQDPSTQAST ASSPAPEE A endothelium, binds to PSEGQRVWGQ GQSPRPENSL CX3CR1.
EREEMGPVPA HTDAFQDWGP GSMAHVSWP VSSEGTPSRE PVASGSWTPK AEEPIHATMD PQRLGVLITP VPDAQAATRR QAVGLLAFLG LLFCLGVAMF TYQSLQGCPR KMAGEMAEGL RYIPRSCGSN SYVLVPV CCL7 (small QPVGINT STTCCYRFIN Chemotactic factor that inducible cytokine KKIPKQRLES YRRTTSSHCP attracts monocytes and A7) REAVIFKTKL DKEICADPTQ eosinophils, but not KWVQDFMKHL DKKTQTPKL neutrophils. Augments Swiss-Prot: P80098 monocyte anti-tumor activity.
Also induces the release of gelatinase B. This protein can bind heparin. Binds to CCR1, CCR2 and CCR3. 60 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 61 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 3 5-324).
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 62 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, 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. 63 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.
By administering a QC (EC)-inhibitor and/or a combination according to the present invention to a mammal it can be possible to prevent or alleviate or treat conditions selected from Alzheimer's disease, Down Syndrome, ulcer disease and gastric cancer with or w/o Helicobacter pylori infections, neoplasia, inflammatory host responses, cancer, melanoma, malign metastasis, psoriasis, rheumatoid arthritis, atherosclerosis, leukocyte adhesion and migration processes in the endothelium, impaired food intake, sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance and impaired regulation of body fluids. 64 Furthermore, by administration of a QC (EC)-inhibitor and/or a combination 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 (EC)-inhibitor and/or a combination according to the present invention can lead to suppression of male fertility.
In a preferred embodiment, the present invention provides a composition, preferably a pharmaceutical composition comprising at least one QC (EC) inhibitor of formula 1 optionally in combination with at least one compound selected from the group consisting of PEP-inhibitors, LiCI, inhibitors of dipeptidyl aminopeptidases, preferably inhibitors of DP IV or DP IV-like enzymes, NPY-receptor ligands, NPY agonists, acetylcholinesterase (ACE) inhibitors, PIMT enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of neutral endopeptidase, inhibitors of Phosphodiesterase-4 (PDE-4), monoamine oxidase (MAO) inhibitors, TNFalpha inhibitors, amyloid protein or amyloid peptide deposition inhibitors, sigma-1 receptor inhibitors and histamine H3 antagonists.
Further, the present invention provides pharmaceutical compositions e.g. for parenteral, enteral or oral administration, comprising at least one QC inhibitor of formula 1 optionally in combination with at least one compound selected from the group consisting of PEP-inhibitors, LiCI, inhibitors of dipeptidyl aminopeptidases, preferably inhibitors of DP IV or DP IV-like enzymes, NPY-receptor ligands, NPY agonists, acetylcholinesterase (ACE) inhibitors, protein isoaspartate carboxymethyl transferase (PIMT) enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of neutral endopeptidase, inhibitors of Phosphodiesterase-4 (PDE-4), MAO inhibitors, TNFalpha inhibitors, amyloid protein or amyloid peptide deposition inhibitors, sigma-1 receptor inhibitors and histamine H3 antagonists, optionally in combination with customary carriers and/or excipients.
These combinations provide a particularly beneficial effect on behavioral conditions and such combinations are therefore shown to be effective and useful for the treatment of neuronal disorders, e.g. neuronal diseases selected from the group consisting of Alzheimer's disease, Down Syndrome, Parkinson disease, Chorea 65 Huntington, pathogenic psychotic conditions, schizophrenia, impaired food intake, sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance, impaired regulation, body fluids, hypertension, fever, sleep dysregulation, anorexia, anxiety related disorders including depression, seizures including epilepsy, drug withdrawal and alcoholism, neurodegenerative disorders including cognitive dysfunction and dementia.
Accordingly, the invention provides a method for the treatment of neuronal disorders, e.g. neuronal diseases selected from the group consisting of Alzheimer's disease, Down Syndrome, Parkinson disease, Chorea Huntington, pathogenic psychotic conditions, schizophrenia, impaired food intake, sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance, impaired regulation, body fluids, hypertension, fever, sleep dysregulation, anorexia, anxiety related disorders including depression, seizures including epilepsy, drug withdrawal and alcoholism, neurodegenerative disorders including cognitive dysfunction and dementia, which comprises administering of a thereutically effective amount said compositions or combinations to a mammal, preferably a human..
Accordingly, the invention provides the use of these compositions or combinations for the preparation of a medicament for the treatment of neuronal disorders, e.g. neuronal diseases selected from the group consisting of Alzheimer's disease, Down Syndrome, Parkinson disease, Chorea Huntington, pathogenic psychotic conditions, schizophrenia, impaired food intake, sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance, impaired regulation, body fluids, hypertension, fever, sleep dysregulation, anorexia, anxiety related disorders including depression, seizures including epilepsy, drug withdrawal and alcoholism, neurodegenerative disorders including cognitive dysfunction and dementia.
The method comprises either co-administration of at least one QC inhibitor of formula 1 and at least one compound selected from the group consisting of PEP-inhibitors, LiCI, inhibitors of dipeptidyl aminopeptidases, preferably inhibitors of DP IV or DP IV-like enzymes, NPY-receptor ligands, NPY agonists, ACE inhibitors, PIMT enhancers, 66 inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of neutral endopeptidase, inhibitors of PDE-4, MAO inhibitors, TNFalpha inhibitors, amyloid protein or amyloid peptide deposition inhibitors, sigma-1 receptor inhibitors and histamine H3 antagonists or the sequential administration thereof.
Co-administration includes administration of a formulation which includes at least one QC inhibitor of formula 1 and at least one compound selected -from the group consisting of PEP-inhibitors, LiCI, inhibitors of dipeptidyl aminopeptidases, preferably inhibitors of DP IV or DP IV-like enzymes, NPY-receptor ligands, NPV agonists, ACE inhibitors, PIMT enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of neutral endopeptidase, inhibitors of PDE-4, MAO inhibitors, TNFalpha inhibitors, amyloid protein or amyloid peptide deposition inhibitors, sigma-1 receptor inhibitors and histamine H3 antagonists or the essentially simultaneous administration of separate formulations of each agent.
Examples of suitable PIMT enhancers are 10-aminoaliphatyl-dibenz[b, f] oxepines of the general formula described in WO 98/15647 and WO 03/057204, respectively, wherein alk is a divalent aliphatic radical, R is an amino group that is unsubstituted or mono- or di-substituted by monovalent aliphatic and/or araliphatic radicals or disubstituted by divalent aliphatic radicals, and Ri, R2, R3 and R4 are each, independently of the others, hydrogen, lower alkyl, lower alkoixy, halogen or trifluoromethyl.
Further useful according to the present invention are modulators of PIMT activity of the general formulae I - IV: wherein the definition of the substituents R1 - R5, (R3)p, (R6)p, X, Y and Z is described in WO 2004/039773.
WO 98/15647, WO 03/057204 and WO 2004/039773 are incorporated herein in their entirety and are part of this invention with regard to the synthesis and use of the compounds described therein.
Suitable inhibitors of beta and/or gamma secretases and compositions containing such inhibitors are described, e.g. in GB 2 385 124, GB 2 389 113, US 2002-115616, WO 01/87293, WO 03/057165, WO 2004/052348 and WO 2004/062652. These references are incorporated herein in their entirety and are part of this invention with regard to the synthesis, manufacture and use of the compounds and compositions described therein for the inhibition of beta and/or gamma secretases.
A potent selective and cell permeable gamma secretase inhibitor is (5S)-(t-Butoxycarbonylamino)-6-phenyl-(4R)hydroxy-(2R)benzylhexanoyl)-L-leu-L-phe-amide with the formula: A potent beta secretase inhibitor is PNU-33312 of the formula: Suitable PDE-4 inhibitors are, e.g. shown in the table below: A preferred PDE-4-inhibitor is Rolipram.
A suitable MAO-inhibitor is the compound ladostigil of the formula 71 Suitable histamine H3 antagonists are, e.g. shown in the table below: Suitable inhibitors of prolyl endopeptidase (PEP) are, e.g. chemical derivatives of proline or small peptides containing terminal prolines. Benzyloxycarbonyl-prolyl- prolinal has been shown to be a specific transition state inhibitor of the enzyme (Wilk, S. and Orloeski, M., J. Neurochem., 41 , 69 (1983), Friedman, et al., Neurochem., 42, 237 (1984)). N-terminal substitutions of L-proline or L-prolylpyrrolidine (Atack, et al., Eur. J. of Pharm., 205, 157-163 (1991), JP 03 56,460, EP 384,341), as well as variations of N-benzyloxycarbonyl (Z) dipeptides containing prolinal at the carboxy terminus have been synthesized as prolyl endopeptidase inhibitors (Nishikata, et al., Chem. Pharm. Bull. 34(7), 2931-2936 (1986), Baker, A. et al., Bioorganic & Medicinal Chem. Letts., 1 (11), 585-590 (1991)). Thioproline, thiazolidine, and oxopyrrolidine substitutions of the core structure have been reported to inhibit prolyl endopeptidase (Tsuru, et al., J. Biochem., 94, 1179 (1988), Tsuru, et al., J. Biochem., 104, 580-586 (1988), Saito et al., J. Enz. Inhib. 5, 51-75 (1991), Uchida, I., et al. PCT Int. Appl. WO 90 12,005, JP 03 56,461 , JP 03 56,462). Similarly, various modifications of the carboxy terminal proline have been made, including various fluorinated ketone derivatives (Henning, EP 4,912,127). General syntheses of fluorinated ketone derivatives has been described (Angelastro, M.R., et al., Tetrahedron Letters 33(23), 3265-3268 (1992)). Other compounds such as chloromethyl ketone derivatives of acyl-proline or acylpeptide-proline (Z-Gly-Pro-CH2CI) have been demonstrated to inhibit the enzyme by alkylating the enzyme's active site (Yoshimoto, T., et al., Biochemistry 16, 2942 (1977)). 74 EP-A-0 286 928 discloses 2-acylpyrroIidine derivatives useful as propyl endopeptidase inhibitors.
Further suitable prolyl endopeptidase inhibitors according to the present invention are, e.g. Fmoc-Ala-Pyrr-CN and those listed below: JTP-4819 S-17092 Japan Tobacco Inc Servier (S)-2-{[(S). (hydroxyacatyl)- (2S, 3aS, 7aS)-1{[(R,R)-2- 1-pyrrolidinyl] carbonyl}-N- phenylcyclopropyl] (phenylmethyl)-l -pyrrolidin- carbonyl}-2-[(thiazolidin-3- carboxamid yl)carbonyl] octahydro-7H- indol 75 Further suitable prolyl endopeptidase inhibitors according to the present invention are disclosed in JP 01042465, JP 03031298, JP 04208299, WO 0071144, US 5847155; JP 09040693, JP 10077300, JP 05331072, JP 05015314, WO 9515310, WO 9300361 , EP 0556482, JP 06234693, JP 01068396, EP 0709373, US 5965556, US 5756763, US 6121311, JP 63264454, JP 64000069, JP 63162672, EP 0268190, EP 0277588, EP 0275482, US 4977180, US 5091406, US 4983624, US 5112847, US 5100904, US 5254550, US 5262431 , US 5340832, US 4956380, EP 0303434, JP 03056486, JP 01143897, JP 1226880, EP 0280956, US 4857537, EP 0461677, EP 0345428, JP 02275858, US 5506256, JP 06192298, EP 0618193, JP 03255080, EP 0468469, US 5118811, JP 05025125, WO 9313065, JP 05201970, WO 9412474, EP 0670309, EP 0451547, JP 06339390, US 5073549, US 4999349, EP 0268281 , US 4743616, EP 0232849, EP 0224272, JP 62114978, JP 62114957, US 4757083, US 4810721, US 5198458, US 4826870, EP 0201742, EP 0201741, US 4873342, EP 0172458, JP 61037764, EP 0201743, US 4772587, EP 0372484, US 5028604, WO 9118877, JP 04009367, JP 04235162, US 5407950, WO 9501352, JP 01250370, JP 02207070, US 5221752, EP 0468339, JP 04211648 and WO 9946272, the teachings of which are herein incorporated by reference in their entirety, especially concerning these inhibitors, their definition, uses and their production.
Most preferred is the PEP-inhibitor of the formula: Other suitable compounds that can be used according to the present invention in combination with QC-inhibitors are NPY, a NPY mimetic or a NPY agonist or antagonist or a ligand of the NPY receptors.
Preferred according to the present invention are antagonists of the NPY receptors. 76 Suitable ligands or antagonists of the NPY receptors are 3a,4,5,9b-tetrahydro-1h-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 0 614 911 , EP 0 747 357, EP 0 747 356 and EP 0 747 378; international patent applications WO 9417035, WO 9719911 , WO 9719913, WO 9612489, WO 9719914, WO 9622305, WO 9640660, WO 9612490, WO 9709308, WO 9720820, WO 9720821 , WO 9720822, WO 9720823, WO 9719682, WO 9725041 , WO 9734843, WO 9746250, WO 9803492, WO 9803493, WO 9803494 and WO 9807420; WO 0030674, US patents Nos. 5552411 , 5663192 and 5567714; 6114336, Japanese patent application JP 09157253; international patent applications WO 9400486, WO 9312139, WO 9500161 and WO 9915498; US Patent No. 5328899; German patent application DE 393 97 97; European patent applications EP 355 794 and EP 355 793; and Japanese patent applications JP 06116284 and JP 07267988, the disclosures in all of which documents are hereby incorporated by reference. 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 0 614 911 , EP 0 747 357, EP 0 747 356 and EP 0 747 378; international patent applications WO 9417035, WO 9719911 , WO 9719913, WO 9612489, WO 9719914, WO 9622305, WO 9640660, WO 9612490, WO 9709308, WO 9720820, WO 9720821 , WO 9720822, WO 9720823, WO 9719682, WO 9725041 , WO 9734843, WO 9746250, WO 9803492, WO 9803493, WO 9803494, WO 9807420 and WO 9915498 ; US patents Nos. 5552411 , 5663192 and 5567714; 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 9417035, WO 9719911 , WO 9719913, WO 9719914 or, preferably, WO 9915498. Preferred amino acid-based NPY antagonists include those that are specifically disclosed in these patent documents, for example 77 B1BP3226 and, especially, (R)-N2-(diphenylacetyl)-(R)-N-[1-(4-hydroxy- phenyl) ethyl] arginine amide (Example 4 of international patent application WO 9915498).
For the avoidance of doubt, the examples disclosed in each of the above mentioned publications are specifically incorporated herein by reference in their entirety, as individually disclosed compounds, especially concerning their structure, their definition, uses and their production.
Suitable DP IV-inhibitors are those, disclosed e.g. in US 6380398, US 6011155; US 6107317; US 6110949; US 6124305; US 6172081 ; WO 9515309, WO 9961431 , WO 9967278, WO 9967279, DE 198 34 591, WO 9740832, DE 196 16 486 C 2, WO 9819998, WO 0007617, WO 9938501 , WO 9946272, WO 9938501 , WO 0168603, WO 0140180, WO 0181337, WO 0181304, WO 0155105, WO 0202560 and WO 0214271, WO 0204610, WO 02051836, WO 02068420, WO 02076450; WO 02083128, WO 0238541 , WO 03000180, WO 03000181 , WO 03000250, WO 03002530, WO 03002531, WO 03002553, WO 03002593, WO 03004496, WO 03004498, WO 03024965, WO 03024942, WO 03035067, WO 03037327, WO 03035057, WO 03045977, WO 03055881 , WO 0368748, WO 0368757, WO 03057666, WO 03057144, WO 03040174, WO 03033524 and WO 03074500, the teachings of which are herein incorporated by reference in their entirety, especially concerning these inhibitors, their definition, uses and their production.
Further suitable DP IV-inhibitors are, e.g. shown in the table below: 78 Company Drug Code Structure Eisai Co Ltd Ferring Research Ltd FE-999011 GlaxoSmithKline pic GW-229A Kyowa Hakko Kogyo K-579 Merck & Co Inc MK-431 Novartis AG LAF-237 N 0 111 80 For the avoidance of doubt, the references and examples disclosed herein are specifically incorporated herein by reference in their entirety, or as individually disclosed compounds, especially concerning their structure, their definition, uses and their production.
Preferred DP IV-inhibitors are dipeptide-like compounds and compounds analogous to dipeptide compounds that are formed from an amino acid and a thiazolidine or pyrrolidine group, and salts thereof, referred to hereinafter as dipeptide-like compounds. Preferably the amino acid and the thiazolidine or pyrrolidine group are bonded with an amide bond.
Especially suitable for that purpose according to the invention are dipeptide-like compounds in which the amino acid is preferably selected from a natural amino acid, such as, for example, leucine, valine, glutamine, glutamic acid, proline, isoleucine, asparagines and aspartic acid.
The dipeptide-like compounds used according to the invention exhibit at a concentration (of dipeptide compounds) of 10 μ , a reduction in the activity of plasma dipeptidyl peptidase IV or DP IV-analogous enzyme activities of at least 0 %, especially of at least 40 %. Frequently a reduction in activity of at least 60 % or at least 70 % is also required. Preferred agents may also exhibit a reduction in activity of a maximum of 20 % or 30 %.
Preferred dipeptide-like compounds are 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.
Further preferred compounds are given in Table 2. 81 The salts of the dipeptide-like compounds can be present in a molar ratio of dipeptide (-analogous) component to salt component of 1 : 1 or 2 : 1. Such a salt is, for example, (lle-Thia)2 fumaric acid.
Table 2: Structures of further preferred dipeptide compounds DP IV-inhibitor H-Asn-pyrrolidine H-Asn-thiazolidine H-Asp-pyrrolidine H-Asp-thiazolidine H-Asp(NHOH)-pyrrolidine H-Asp(NHOH)-thiazolidine H-Glu-pyrrolidine H-Glu-thiazoIidine H-Glu(NHOH)-pyrroiidine H-Glu(NHOH)-thiazolidine H-His-pyrrolidine H-His-thiazolidine H-Pro-pyrrolidine H-Pro-thiazolidine H-lle-azididine H-lle-pyrrolidine H-L-a//o-lle-thiazolidine H-Val-pyrrolidine H-Val-thiazolidine In another preferred embodiment, the present invention provides the use of compounds of formula 3 for competitive modulation of dipeptidyl peptidase IV catalysis: K C E (3) wherein 82 A, B, C, D and E are independently any amino acid moieties including proteinogenic amino acids, non-proteinogenic amino acids, L-amino acids and D-amino acids and wherein E and/or D may be absent.
According to preferred embodiments, the residues A, B, C, D and E of formula (3) are independently defined as follows: A is an amino acid except a D-amino acid, B is an amino acid selected from Pro, Ala, Ser, G\y, Hyp, acetidine-(2)-carboxy!ic acid and pipecolic acid, C is any amino acid except Pro, Hyp, acetidine-(2)-carboxylic acid, pipecolic acid and except N-alkylated amino acids, e.g. N-methyl valine and sarcosine, D is any amino acid or missing, and E is any amino acid or missing, or: C is any amino acid except Pro, Hyp, acetidine-(2 carboxylic acid, pipecolic acid, except N-alkylated amino acids, e.g. N-methyl valine and sarcosine, and except a D-amino-acid; D is any amino acid selected from Pro, Ala, Ser, Gly, Hyp, acetidine-(2)- carboxylic acid and pipecolic acid, and E is any amino acid except Pro, Hyp, acetidine-(2>-carboxylic acid, pipecolic acid and except N-alkylated amino acids, e.g. N-methyl valine and sarcosine.
Other amino acids than those encoded in the genetic code can also be included in peptide compounds within the scope of the invention and can be classified within this general scheme.
Proteinogenic amino acids are defined herein as natural protein-derived a-amino acids. Non-proteinogenic amino acids are defined hierein as all other amino acids, which are not building blocks of common natural proteins.
The resulting peptides may be synthesized as the free C-terminal acid or as the C-terminal amide form. The free acid peptides or the amides may be varied by side 83 chain modifications. Such side chain modifications include for instance, but are not restricted to, homoserine formation, pyroglutamic acid formation, disulphide bond formation, deamidation of asparagine or glutamine residues, methylation, t-butylation, t-butyloxycarbonylation, 4-methylbenzylation, thioanysilation, thiocresylation, benzyloxymethylation, 4-nitrophenylation, benzyloxycarbonylation, 2-nitrobencoylation, 2-nitrosulphenylation, 4-toluenesulphonylation, pentafluorophenylation, diphenylmethylation, 2-chlorobenzyloxycarbonylation, 2,4,5-trichlorophenylation, 2-bromobenzyloxycarbonylation, 9-fluorenylmethyloxycarbonylation, triphenylmethylation, 2,2,5,7,8,-pentamethylchroman-6-sulphonylation, hydroxylation, oxidation of methionine, formylation, acetylation, anisylation, benzylation, bencoylation, trifluoroacetylation, carboxylation of aspartic acid or glutamic acid, phosphorylation, sulphation, cysteinylation, glycolysation with pentoses, deoxyhexoses, hexosamines, hexoses or N-acetylhexosamines, farnesylation, myristolysation, biotinylation, palmitoylation, stearoylation, geranylgeranylation, glutathionylation, 5'-adenosylation, ADP-ribosylation, modification with N-glycolylneuraminic acid, N-acetylneuraminic acid, pyridoxal phosphate, lipoic acid, 4'-phosphopantetheine, or N-hydroxysuccinimide.
In the compounds of formula (3), the amino acid moieties A, B, C, D, and E are respectively attached to the adjacent moiety by amide bonds in a usual manner according to standard nomenclature so that the amino-terminus (N-terminus) of the amino acids (peptide) is drawn on the left and the carboxyl-terminus of the amino acids (peptide) is drawn on the right. (C-terminus).
Preferred peptide compounds are listed in table 3.
Table 3: Examples of peptide substrates Peptide Mass (calc.) Mass (exp.)1 [M+H*] 2-Amino octanoic acid-Pro-lle 369.5 370.2 Abu-Pro-lle 313.4 314.0 Aib-Pro-lle 313.4 314.0 Aze-Pro-lle 311.4 312.4 Cha-Pro-lle 381.52 382.0 lle-Hyp-lle 356.45 358.2 84 1 [ +hT] were determined by Electrospray mass spectrometry in positive ionization mode. i-butyl-Gly is defined as: 85 Ser(Bzl) and Ser(P) are defined as benzyl-serine and phosphoryl-serine, respectively. Tyr(P) is defined as phosphoryl-tyrosine.
Further preferred DP IV-inhibitors, which can be used according to the present invention, are peptidylketones of formula 4: and pharmaceutically acceptable salts thereof, wherein: A is selected from the following structures: wherein X1 is H or an acyl or oxycarbonyl group including an amino acid residue, N- protected amino acid residue, a peptide residue or a N-protected peptide residue, X2 is H, -(CH)m-NH-C5H3N-Y with m = 2-4 or -C5H3N-Y (a divalent pyridyl residue) and Y is selected from H, Br, CI, I, NO2 or CN, X3 is H or selected from an alkyl-, aikoxy-, halogen-, nitro-, cyano- or carboxy- substituted phenyl or from an alkyl-, aikoxy-, halogen-, nitro-, cyano- or carboxy- substituted pyridyl residue, X4 is H or selected from an alkyl-, aikoxy-, halogen-, nitro-, cyano- or carboxy- substituted phenyl or from an alkyl-, aikoxy-, halogen-, nitro-, cyano- or 86 carboxy- substituted pyridyl residue, X5 is H or an alkyl, alkoxy or phenyl residue, Xs is H or an alkyl residue, for n = 1 X is selected from: H, OR2, SR2, NR R3, N+R2R3R4, wherein: R2 stands for acyl residues, which are optionally substituted with alkyl, cycloalkyi, aryl or heteroaryl residues, or for amino acid residues or peptidic residues, or alkyl residues, which are optionally substituted with alkyl, cycloalkyi, aryl or heteroaryl residues, R3 stands for alkyl or acyl residues, wherein R2 and R3 may be part of a saturated or unsaturated carbocyclic or heterocyclic ring, R4 stands for alkyl residues, wherein R2 and R4 or R3 and R4 may be part of a saturated or unsaturated carbocyclic or heterocyclic ring, for n = 0 X is selected from: wherein B stands for: O, S or NR5, wherein R5 is H, alkyl or acyl, C, D, E, F, G, Y, K, L, M, Q, T, U, V and W are independently selected from alkyl and substituted alkyl residues, oxyalkyl, thioalkyl, aminoalkyl, carbonylalkyl, acyl, carbamoyl, aryl and heteroaryl residues, and Z is selected from H, or a branched or straight chain alkyl residue from C1-C9, a branched or straight chain alkenyl residue from C2-C9, a cycloalkyi residue from C3-C8, a cycloalkenyl residue from C5-C7, an aryl or heteroaryl residue, or a side chain selected from all side chains of all natural amino acids or derivatives thereof. 87 In preferred compounds of formula 4, A is X1 N' X2 wherein X1 is H or an acyl or oxycarbonyl group including an amino acid residue, N- acylated amino acid residue, a peptide residue from d - to pentapeptides, preferably a dipeptide residue, or a N-protected peptide residue from di- to pentapeptides, preferably a N-protected dipeptide residue X2 is H, -(CH)m-NH-C5H3N-Y with m = 2-4 or -C5H3N-Y (a divalent pyridyl residue) and Y is selected from H, Br, CI, I, N02 or CN, for n = 1 X is preferably selected from: H, OR2, SR2, NR2R3, wherein: R2 stands for acyl residues, which are optionally substituted with alkyl, cycloalkyi, aryl or heteroaryl residues, or for amino acid residues or peptidic residues, or alkyl residues, which are optionally substituted with alkyl, cycloalkyi, aryl or heteroaryl residues, R3 stands for alkyl or acyl residues, wherein R2 and R3 may be part of a saturated or unsaturated carbocyclic or heterocyclic ring, for n = 0 preferably selected from: wherein B stands for: O, S or NR5, wherein R5 is H, alkyl or acyl, C, D, E, F, G, Y, K, L, M and Q are independently selected from alkyl and substituted alkyl residues, oxyalkyi, thioalkyi, aminoalkyi, carbonylalkyi, acyl, carbamoyl, aryl and heteroaryl residues, and 88 Z is selected from H, or a branched or straight chain alkyl residue from (VCg, preferably C2- C6l a branched or straight chain alkenyl residue from C2-C9, a cycloalkyi residue from C3-C8, a cycloalkenyl residue from C5-C7, an aryl or heteroaryl residue, or a side chain selected from all side chains of all natural amino acids or derivatives thereof.
In more preferred compounds of formula 4, A is wherein X1 is H or an acyl or oxycarbonyl group including an amino acid residue, N- acylated amino acid residue or a peptide residue from di- to pentapeptides, preferably a dipeptide residue, or a N-protected peptide residue from di- to pentapeptides, preferably a N-protected dipeptide residue for n = 1 , X is preferably selected from: H, OR2, SR2, wherein: R2 stands for acyl residues, which are optionally substituted with alkyl or aryl residues, for n = 0 preferably selected from: wherein B stands for: O, S or NR5, wherein R5 is H, alkyl or acyl, C, D, E, F, G, Y, K, L, M and Q are independently selected from alkyl and 89 substituted alkyl residues, oxyalkyi, thioalkyi, aminoalkyl, carbonylalkyl, acyl, carbamoyl, aryl and heteroaryl residues, and Z is selected from H, or a branched or straight chain alkyl residue from C-1-C9, preferably C2- C6, a branched or straight chain alkenyl residue from C2-C9, a cycloalkyi residue from C3-CB) a cycloalkenyl residue from C5-C7, an aryl or heteroaryl residue, or a side chain selected from all side chains of all natural amino acids or derivatives thereof.
In most preferred compounds of formula 4, A is wherein X1 is H or an acyl or oxycarbonyl group including an amino acid residue, N- acylated amino acid residue or a dipeptide residue, containing a Pro or Ala in the penultimate position, or a N-protected dipeptide residue containing a Pro or Ala in the penultimate position, for n = 1 , X is H, for n = 0 X is preferably selected from: wherein B stands for: O or S, most preferably for S C, D, E, F, G, Y, K, L, M, Q, are H and Z is selected from H, or a branched or straight chain alkyl residue from C3-C5, a 90 branched or straight chain alkenyl residue from C2-C9, a cycloalkyl residue from C5-C7, a cycloalkenyl residue from C5-C7, an aryl or heteroaryl residue, or a side chain selected from all side chains of all natural amino acids or derivatives thereof.
Most preferred for Z is H.
According to a preferred embodiment the acyl groups are CrC6-acyl groups.
According to a further preferred embodiment the alk(yl) groups are Ci-C6-alk(yI) groups, which may be branched or unbranched.
According to a still further preferred embodiment the alkoxy groups are Ci-C3-alkoxy groups.
According to yet another preferred embodiment the aryl residues are C5-C12 aryl residues that have optionally fused rings.
According to a still further preferred embodiment the cycloalkyl residues (carbocycles) are C3-C8-cycloalkyl residues.
According to another preferred embodiment the heteroaryl residues are C4-Cn aryl residues that have optionally fused rings and, in at least one ring, additionally from 1 to 4 preferably 1 or 2 hetero atoms, such as O, N and/or S.
According to a further preferred embodiment peptide residues contain from 2 to 50 amino acids.
According to another preferred embodiment the heterocyclic residues are C2-C7-cycloalkyl radicals that additionally have from 1 to 4, preferably 1 or 2 hetero atoms, such as O, N and/or S.
According to a still further preferred embodiment the carboxy groups are Ci - C6 carboxy groups, which may be branched or unbranched.
According to yet another preferred embodiment the oxycarbonyl groups are groups of the formula -0-(CH2)i-6COOH.
The amino acids can be any natural or synthetic amino acid, preferably natural alpha amino acids.
Preferred compounds of formula (4) are 2-Methylcarbonyl-1-N-[(L)-Alanyl-(L)-Valinyl]-(2S)-pyrrolidine hydrobromide; 2- ethyl)carbonyl-1-N-[(L)-Valinyl-(L)-Prolyl-(L)-Valinyl]-(2S)-pyrroIidine hydrobromide; 2-[(Acetyl-oxy-methyl)carbonyl]-1 -N-[(L)-Alanyl-(L)-VaIinyl]-(2S)-pyrrolidine hydrobromide; 2-[Benzoyl-oxy-methyl)carbonyl]-1 -N-[{(L)-Alanyl}-(L)-Valinyl]-(2S)-pyrrolidine hydrobromide; 2-{[(2,6- 91 Dichlorbenzy thiomet^ 2-[Benzoy-Ioxy-methyl)carbonyl]-1-N-[Glycyl-(L)-Valinyl]-(2S)-pyrrolidine hydrobromide; 2-[([1,3]-thiazole-2-yl)carbonyl]-1-N-[{(L)-Alanyl}-(L)-Valinyl]-(2S)-pyrro!idine trifluoracetat; 2-[(benzothiazoIe-2-yl)carbonyl]-1-N-[N-{(L)-AlanylHL)-Valinyl]-(2S)-pyrrolidin trifluoracetat; 2-[(-benzothiazole-2-yl)carbonyl]-1-N-[{(L)-Alanyl}-Glycyl]-(2S)-pyrrolidine trifluoracetat; 2-[(pyridin-2-yl)carbonyl]-1 -N-[N-{(L)-Alanyl}-(L)-Valinyl]-(2S)-pyrrolidine trifluoracetat.
Further, according to the present invention preferred DP IV-inhibitors are compounds of formula (5) including all stereoisomers and pharmaceutical acceptable salts: B— (CH~R )n-C(=X2)— D (5) wherein n is 0 or , R1 stands for H, C-1-C9 branched or straight chain alkyl, preferably H, n-butan-2-yl, n-prop-2-yl or isobutyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, preferably cyclohexyl, C5-C7 cycloalkenyl, aryl, heteroaryl or a side chain of a natural amino acid or mimetics thereof, X2 stands for O, NR6, N+(R7)2, or S, B is selected from the following groups: where X5 is H or an acyl or oxycarbonyl group including amino acids, is H, C^Cg branched or straight chain alkyl, preferably H, n-butan-2-yl, n- prop-2-yl or isobutyl, C2-C9 branched or straight chain alkenyl, C3-C8 cycloalkyl, preferably cyclohexyl, 3-hydroxyadamant-1-yl, C5-C7 cycloalkenyl, aryl, heteroaryl or a side chain of a natural amino acid or derivatives thereof, or a group of the formula -(CH)m-NH-C5H3N-Y where m is an integer of 2-4, - 92 C5H3N-Y is a divalent pyridyl moiety and Y is a hydrogen atom, a halogen atom, a nitro group or a cyano group, R6, R7 and R8 are independently selected from H, optionally substituted C1-C9 branched or straight chain alkyl, preferably an optionally substituted C2-C5 branched or straight chain alkyl; or optionally substituted C2-C9 branched or straight chain alkenyl, preferably an C2-C5 branched or straight chain alkenyl; or optionally substituted C3-C8 cycloalkyi, preferably an optionally substituted C4-C7 cycloalkyi; or an optionally substituted C5-C7 cycloalkenyl, or an optionally substituted aryl residue, Z is selected from H, pyridyl or optionally substituted phenyl, optionally substituted alkyl groups, alkoxy groups, halogens, nitro, cyano and carboxy groups, W is selected from H, pyridyl or optionally substituted phenyl, optionally substituted alkyl groups, alkoxy groups, halogens, nitro, cyano and carboxy groups, W1 is H or optionally substituted alkyl, alkoxy or optionally substituted phenyl, and Z1 is H, or optionally substituted alkyl, R3 and R4 are independently H, hydroxy, alkyl, alkoxy, aralkoxy, nitro, cyano or halogen, is an optionally substituted compound of the formula can be saturated, or can have one, two or three double bonds, X8 to X11 are independently CH, N, N+(R7), or CR8, if unsaturated, or X8 to X1 are independently CH2, NH, NH+(R7), O, or S if saturated, X12 is CHA, NA, CH2, NH, NH+(R7), or CHR8, if saturated or 93 X12 is CA, A+, CH, N, N+(R7), or CR8, if unsaturated and A is H or an isoster of a carboxylic acid such as CN, S03H, CONOH, P03R5Re, a tetrazole, an amide, an ester or an acid anhydride.
Throughout the application, D contains preferably at most two, further preferred at most one hetero atom in the ring.
According to preferred embodiments of the present invention, D stands for optionally substituted C4-C7 cycloalkyi, preferably C4-C6 cycloalkyi, optionally substituted C4-C7 cycloalkenyl, or optionally substituted (hetero)cycloalkyl of the formulae wherein the residues are as defined above, or that is, a five-membered ring containing one or two double bonds in the ring, wherein the residues are as defined above, or wherein the residues are as defined above, or wherein the residues are as defined above, 94 or that is a six-membered ring containing one or two double bonds in the ring, wherein the residues are as defined above, or wherein the residues are as defined above.
According to a preferred embodiment, B has the following formula: wherein the residues are as defined above. - According to another preferred embodiment, B has the following formula: wherein the residues are as defined above.
Preferred compounds according to formula (5) are 1-cyclopentyl-3-methyl-1-oxo-2-pentanaminium chloride, i-cyclopentyl-3-methyl-1 -oxo-2-butanaminium chloride, 1 -cyclopentyl-3,3-dimethyl-1 -oxo-2-butanaminium chloride, 1 -cyclohexyl-3,3-dimethyl-1 -oxo-2-butanaminium chloride, 95 3-(cyclopentylcarbonyl)-1 ,2,3,4-tetrahydroisoquinolinium chloride, and N-(2-cyclopentyl-2-oxoethyl)cyclohexanaminium chloride.
Because of the wide distribution of the protein in the body and the wide variety of mechanisms involving DP IV, DP IV-activity and DP IV-related proteins, systemic therapy (enteral or parenteral administration) with DP IV-inhibitors can result in a series of undesirable side-effects.
The problem to be solved was therefore moreover, to provide DP IV-inhibitors that can be used in combination therapy of neuronal diseases, for targeted influencing of locally limited patho-physiological and physiological processes. The problem of the invention especially consists in obtaining locally limited and highly specific inhibition of DP IV or DP IV-analogous activity for the purpose of targeted intervention in the regulation of the activity of locally active substrates.
This problem is solved according to the invention by the use of the DP IV-inhibitors of the general formula (6): B (6) wherein A is an amino acid having at least one functional group in the side chain, B is a chemical compound covalently bound to at least one functional group of the side chain of A, C is a thiazolidine, pyrrolidine, cyanopyrrolidine, hydroxyproline, dehydroproline piperidine group amide-bonded to A.
In accordance with a preferred embodiment of the invention, pharmaceutical compositions are used comprising at least one compound of the general formula (5) and at least one customary adjuvant appropriate for the site of action. 96 Preferably A is an a-amino acid, especially a natural α-amino acid having one, two or more functional groups in the side chain, preferably threonine, tyrosine, serine, arginine, lysine, aspartic acid, glutamic acid or cysteine.
Preferably B is an oligopeptide having a chain length of up to 20 amino acids, a polyethylene glycol having a molar mass of up to 20 000 g/mol, an optionally substituted organic amine, amide, alcohol, acid or aromatic compound having from 8 to 50 C atoms.
Despite an extended side chain function, the compounds of formula (6) can still bind to the active centre of the enzyme dipeptidyl peptidase IV and analogous enzymes but are no longer actively transported by the peptide transporter PepT1. The resulting reduced or greatly restricted transportability of the compounds according to the invention leads to local or site directed inhibition of DP IV and DP IV-like enzyme activity.
By extending/expanding the side chain modifications, for example beyond a number of seven carbon atoms, it is accordingly possible to obtain a dramatic reduction in transportability. With increasing spatial size of the side chains, there is a reduction in the transportability of the substances. By spatially and sterically expanding the side chains, for example beyond the atom group size of a monosubstituted phenyl radical, hydroxylamine radical or amino acid residue, it is possible according to the invention to modify or suppress the transportability of the target substances.
Preferred compounds of formula (6) are compounds, wherein the oligopeptides have chain lengths of from 3 to 15, especially from 4 to 10, amino acids, and/or the polyethylene glycols have molar masses of at least 250 g/mol, preferably of at least 1500 g/mol and up to 15 000 g/mol, and/or the optionally substituted organic amines, amides, alcohols, acids or aromatic compounds have at least 12 C atoms and preferably up to 30 C atoms.
Pharmaceutical compositions To prepare the pharmaceutical compositions of this invention, at least one effector of QC optionally in combination with at least one PEP-in ibitor and/or at least one DP 97 IV-inhibitor and/or at least one NPY-receptor-ligand and/or at least one ACE-inhibitor, 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 98 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 com 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 preform ulation 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 ingred ient 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 99 for aqueous suspensions, include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone or gelatin.
Where the processes for the preparation of the compounds of the present invention give rise to 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-toluoyM-tartaric acid followed by fractional crystallization and regeneration of the free base. The compounds may also 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.
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. . Wilts, Protective Groups in Organic Synthesis. John Wiley & Sons, 1991. The protecting groups may be removed at a convenient subsequent stage using conventional methods known from the art.
The method of treating neuronal disorders as described in the present invention, may also be carried out using a pharmaceutical composition comprising at least one effector of QC optionally in combination with at least one PEP-inhibitor and/or at least one DP IV-inhibitor and/or at least one NPY-receptor-ligand and/or at least one ACE-inhibitor or any other of the compounds as defined herein and a pharmaceutically acceptable carrier. The pharmaceutical composition may contain between about O.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 100 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. 101 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 eneoxidepotyllysine 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, poiyorthoesters, 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 102 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.
Suitably, the particularly beneficial effect provided by the treatment of the invention is an improved therapeutic ratio for the combination of the invention relative to the therapeutic ratio for one compound of the combination when used alone and at a dose providing an equivalent efficacy to the combination of the invention.
In a preferred aspect, the particularly beneficial effect provided by the treatment of the invention is indicated to be a synergistic effect relative to the control expected from the effects of the individual active agents.
In a further aspect of the invention, combining doses of at least one QC-inhibitor with at least one PEP-inhibitor and/or at least one DP IV-inhibitor and/or at least one NPY-receptor-ligand will produce a greater beneficial effect than can be achieved for either agent alone at a dose twice that used for that agent in the combination.
In a preferred aspect, the dosage level of each of the active agents when used in accordance with the treatment of the invention will be less than would have been required from a purely additive effect upon the neuronal condition.
It is also considered that the treatment of the invention will effect an improvement, relative to the individual agents, in decreasing the intracellular deposition of pGlu-amyloid- -peptides and thereby dramatically slowing down the plaque formation in the brain of a mammal, preferably in human brain.
In a further aspect, the invention also provides a process for preparing a pharmaceutical composition comprising at least one at least one effector of QC optionally in combination with at least one PEP-inhibitor and/or at least one DP IV-inhibitor and/or at least one NPY-receptor-ligand and/or at least one ACE-inhibitor and a pharmaceutically acceptable carrier therefor, which process comprises admixing the QC effector and/or DP IV-inhibitor and/or the PEP-inhibitor and/or the 103 NPY-receptor-ligand and/or the ACE-inhibitor 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 QC-inhibitor, the PEP-inhibitor, the DP IV-inhibitor and the NPY-receptor-ligand 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 31st Edition page 341 and pages cited therein) or the above mentioned publications.
Examples of the invention Example 1: Solid-phase synthesis of peptides The peptides used herein were synthesized with an automated synthesizer SYMPHONY (RAININ) using a modified Fmoc-protocol. Cycles were modified by using double couplings from the 15th amino acid from the C-terminus of the peptide with five-fold excess of Fmoc-amino acids and coupling reagent. The peptide couplings were performed by TBTU/NMM-activation using a 0.23 mmol substituted NovaSyn TGR-resin or the corresponding preloaded Wang-resin at 25 mohscale. The cleavage from the resin was carried out by a cleavage-cocktail consisting of 94.5 % TFA, 2.5 % water, 2.5 % EDT and 1 % TIS.
Analytical and preparative HPLC were performed by using different gradients on the LiChrograph HPLC system of Merck-Hitachi. The gradients were made up from two solvents: (A) 0.1 % TFA in H2O and (B) 0.1 % TFA in acetonitrile. Analytical HPLC were performed under the following conditions: solvents were run (1 ml/min) through a 125-4 Nucleosil RP18-column, over a gradient from 5 % - 50 % B over 15 min and then up to 95 % B until 20 min, with UV detection (λ = 220 nm). Purification of the peptides was carried out by preparative HPLC on either a 250-20 Nucleosil 100 RP8- 104 column or a 250-10 LiChrospher 300 RP18-column (flow rate 6 ml/min, 220 nm) under various conditions depending on peptide chain length.
For the identification of the peptides and peptide analogues, laser desorption mass spectrometry was employed using the HP G2025 MALDI-TOF system of Hewlett-Packard.
Example 2: Determination of IC50-values of DP IV~inhibitors 100 μΙ inhibitor stock solution were mixed with 100 μΙ buffer (HEPES pH 7.6) and 50 μΙ substrate (Gly-Pro-pNA, final concentration 0.4 m ) and preincubated at 30°C. Reaction was started by addition of 20 μΙ purified porcine DP IV. Formation of the product pNA was measured at 405 nm over 10 min using the HTS 7000Plus plate reader (Perkin Elmer) and slopes were calculated. The final inhibitor concentrations ranged between 1 mM and 30 nM.
For calculation of IC50-values GraFit 4.0.13 (Erithacus Software) was used.
Example 3: Determination ofKrvalues of DP IV-inhibitors For determination of the Kj-values DP IV activity was measured in the same way as described in example 2 at final substrate concentrations of 0.05, 0.1 , 0.2, and 0.4 mM and further 7 inhibitor concentrations covering the IC50 concentration. Calculations were performed using the GraFit Software.
Example 4: Prolyl endopeptidase (PEP) enzymatic activity assays The enzymatic activity of PEP was quantified as described recently (Schulz et al., 2002, Modulation of inositol 1 ,4,5-triphosphate concentration by prolyl endopeptidase inhibition. Eur J Biochem 269: 5813-5820). Cellular extracts as described above were incubated in the assay buffer using the fluorogenic substrate Z-Gly-Pro-NHMec (10 μΜ; Bachem, Heidelberg, Germany) on a spectrofluorimeter SFM 25 (excitation wavelength 380 nm, emission wavelength 460 nm, Kontron, Neufahrn, Germany) equipped with a four-cell changer and controlled by an IBM-compatible personal computer. The data obtained were analyzed with the software FLUCOL (Machleidt et al., 1995). 105 Example 5: Assays for glutaminyl cyclase activity 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- JNA. 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 320/410 nm. The assay reactions were initiated by addition of glutaminyl cyclase. QC activity was determined from a standard curve of y#-naphthylamine under assay conditions. One unit is defined as the amount of QC catalyzing the formation of 1 pmol pGlu- 3NA from H-Gln- JNA per minute under the described conditions.
In a second fluorometric assay, QC was activity was 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 ct-Ketoglutaric acid and 30 U/ml glutamate dehydrogenase in a final volume 106 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 Kj-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.
Example 6: 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 were 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 we used DHAP/DAHC, prepared by solving 30 mg 2',6'-dihydroxyacetophenone (Aldrich) and 44 mg diammonium hydrogen citrate (Fluka) in 1 ml aceton'itrile/0.1% TFA in water (1/1 , v/v). A small volume (« 1 pi) 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. 107 For long-term testing of Glu1-cyclization, Αβ-derived peptides were incubated in 100μΙ 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(3-11)a] or 0.15 mM [A (3-21)a] concentrations, and 0.2 U QC was added all 24 hours. In case of Ap(3-21)a, the assays contained 1 % DMSO. At different times, samples were 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 did 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 benzimidazole or 2 mM 1 ,10-phenanthroline).
The first QC inhibitors are disclosed in WO 200409859. There are no other potent QC inhibitors known in the art. The same holds true for combinations and compositions for the treatment of neuronal diseases comprising QC inhibitors. 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 drugsor 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 above 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.
PBD00038_Seq . ST25. txt SEQUENCE LISTING <110> Probiodrug AG <120> Novel inhibitors of Glutaminyl cyclase <130> PBD 00038/WO <140> PCT/EP 2005/001153 <141> 2005-02-04 <150> US 60/542,133 <151> 2004-02-05 <150> US 60/634,364 <151> 2004-12-08 <150> US 10/838,993 <151> 2004-05-05 <160> 23 <170> Patentln version 3.1 <210> 1 <211> 42 <212> PRT <213> Homo sapiens <400> 1 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu Val His His Gin Lys 1 5 10 15 Leu Val Phe Phe Ala Glu Asp Val Gly Ser Asn Lys Gly Ala lie lie 25 30 Page 1 PBD00038_Seq . ST25. txt Leu Met val Gly Gly val val lie Ala 40 <210> 2 <211> 40 <212> PRT <213> Homo sapiens <400> 2 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu Val His His Gin Lys 1 5 10 15 Leu val Phe Phe Ala Glu Asp val Gly Ser Asn Lys Gly Ala lie lie 25 30 Gly Leu Met val Gly Gly val val 40 <210> 3 <211> 40 <212> PRT <213> Homo sapiens <400> 3 Glu Phe Arg His Asp Ser Gly Tyr Glu Val His His Gin Lys Leu val 1 5 10 15 Phe Phe Ala Glu Asp val Gly Ser Asn Lys Gly Ala lie lie Gly Leu 25 30 Met Val Gly Gly Val Val lie Ala 40 <210> 4 <211> 38 <212> PRT <213> Homo sapiens <400> 4 Glu Phe Arg His Asp Ser Gly Tyr Glu Val His His Gin Lys Leu Val 1 5 10 15 Page 2 PBD00038_Seq. ST25. txt Phe Phe Ala Glu Asp val Gly Ser Asn Lys Gly Ala lie lie Gly Leu 25 30 Met val Gly Gly val val <210> 5 <211> 11 <212> PRT <213> Artificial sequence <220> <223> Synthetic peptide <220> <221> MOD_RES <222> (11) .. (11) <223> AMIDATION <400> 5 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu 1 5 10 <210> 6 <211> 9 <212> PRT <213> Artificial sequence <220> <223> synthetic peptide <220> <221> MOD_RES <222> (9).. (9) <223> AMIDATION <400> 6 Glu Phe Arg His Asp Ser Gly Tyr Glu 1 5 Page 3 PBD00038_Seq . ST25. txt <210> 7 <211> 21 <212> PRT <213> Artificial sequence <220> <223> synthetic peptide <220> <221> MOD_RES <222> (21).. (21) <223> AMIDATION <400> 7 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu Val His His Gin Lys 1 5 10 15 Leu val Phe Phe Ala <210> 8 <211> 19 <212> PRT <213> Artificial sequence <220> <223> synthetic peptide <220> <221> MOD_RES <222> (19) .. (19) <223> AMIDATION <400> 8 Glu Phe Arg His Asp ser Gly Tyr Glu Val His His Gin Lys Leu Val 1 5 10 15 Phe Phe Ala Page 4 PBD00038_Seq . ST25. txt <210> 9 <211> 38 <212> PRT <213> Artificial sequence <220> <223> Synthetic peptide <400> 9 Gin Phe Arg His Asp ser Gly Tyr Glu val His His Gin Lys Leu val 1 5 10 15 Phe Phe Ala Glu Asp val Gly Ser Asn Lys Gly Ala lie lie Gly Leu 25 30 Met Val Gly Gly Val Val <210> 10 <211> 19 <212> PRT <213> Artificial sequence <220> <223> Synthetic peptide <220> <221> MOD_RES <222> (19).. (19) <223> AMIDATION <400> 10 Gin Phe Arg His Asp Ser Gly Tyr Glu val His His Gin Lys Leu 1 5 10 15 Phe Phe Ala <210> 11 <211> 11 <212> PRT Page 5 PBD00038_Seq . ST25. txt <213> Artificial sequence <220> <223> Synthetic peptide <220> <221> MOD_RES <222> (11) .. (11) <223> AMIDATION <400> 11 Asp Ala Gin Phe Arg His Asp Ser Gly Tyr Glu 1 5 10 <210> 12 <211> 9 <212> PRT <213> Artificial sequence <220> <223> Synthetic peptide <220> <221> MOD_RES <222> (9).. (9) <223> AMIDATION <400> 12 Gin Phe Arg His Asp Ser Gly Tyr Glu 1 5 <210> 13 <211> 17 <212> PRT <213> Homo sapiens <220> <221> MOD_RES Page 6 PBD00038_Seq . ST25. txt <222> (17) . . (17) <223> AMIDATION <400> 13 Gin Gly Pro Trp Leu Glu Glu Glu Glu Glu Ala Tyr Gly Trp Met Asp 1 5 10 15 Phe <210> 14 <211> 13 <212> PRT <213> Homo sapiens <400> 14 Gin Leu Tyr Glu Asn Lys Pro Arg Arg Pro Tyr lie Leu 1 5 10 <210> 15 <211> 10 <212> PRT <213> Homo sapiens <220> <221> MOD_RES <222> (10) . . (10) <223> AMIDATION <400> 15 Gin His Trp Ser Tyr Gly Leu Arg Pro Gly 1 5 10 <210> 16 <211> 97 <212> PRT <213> Homo sapiens Page 7 PBD00038_Seq . ST25. txt <400> 16 Gin Pro Lys val Pro Glu Trp Val Asn Thr Pro Ser Thr Cys Cys Leu 1 5 10 15 Lys Tyr Tyr Glu Lys Val Leu Pro Arg Arg Leu val val Gly Tyr Arg 25 30 Lys Ala Leu Asn cys His Leu Pro Ala lie lie Phe val Thr Lys Arg 40 45 Asn Arg Glu val Cys Thr Asn Pro Asn Asp Asp Trp Val Gin Glu Tyr 50 55 60 lie Lys Asp Pro Asn Leu Pro Leu Leu Pro Thr Arg Asn Leu Ser Thr 65 70 75 80 val Lys lie lie Thr Ala Lys Asn Gly Gin Pro Gin Leu Leu Asn ser 85 90 95 Gin <210> 17 <211> 76 <212> PRT <213> Homo sapiens <400> 17 Gin Pro Asp Ser val Ser lie Pro lie Thr Cys Cys Phe Asn val lie 1 5 10 15 Asn Arg Lys lie Pro lie Gin Arg Leu Glu Ser Tyr Thr Arg lie Thr 25 30 Asn lie Gin Cys Pro Lys Glu Ala Val lie Phe Lys Thr Lys Arg Gly 40 45 Lys Glu val Cys Ala Asp Pro Lys Glu Arg Trp val Arg Asp Ser Met 50 55 60 Lys His Leu Asp Gin lie Phe Gin Asn Leu Lys Pro 65 70 75 <210> 18 <211> 76 <212> PRT Page 8 PBD00038_Seq . ST25. txt <213> Homo sapiens <400> 18 Gin Pro Asp Ala lie Asn Ala Pro val Thr cys Cys Tyr Asn Phe Thr 1 5 10 15 Asn Arg Lys lie Ser Val Gin Arg Leu Ala ser Tyr Arg Arg lie Thr 25 30 Ser Ser Lys Cys Pro Lys Glu Ala Val lie Phe Lys Thr lie Val Ala 40 45 Lys Glu lie Cys Ala Asp Pro Lys Gin Lys Trp val Gin Asp Ser Met 50 55 60 Asp His Leu Asp Lys Gin Thr Gin Thr Pro Lys Thr 65 70 75 <210> 19 <211> 68 <212> PRT <213> Homo sapiens <400> 19 Gin val Gly Thr Asn Lys Glu Leu Cys Cys Leu val Tyr Thr Ser Trp 1 5 10 15 Gin lie Pro Gin Lys Phe lie val Asp Tyr Ser Glu Thr ser Pro Gin 25 30 Cys Pro Lys Pro Gly val lie Leu Leu Thr Lys Arg Gly Arg Gin lie 40 45 Cys Ala Asp Pro Asn Lys Lys Trp Val Gin Lys Tyr lie Ser Asp Leu 50 55 60 Lys Leu Asn Ala 65 <210> 20 <211> 373 <212> PRT <213> Homo sapiens Page 9 PBD00038_Seq . ST25. txt <400> 20 Gin His His Gly Val Thr Lys Cys Asn lie Thr Cys Ser Lys Met Thr 1 5 10 15 Ser Lys lie Pro Val Ala Leu Leu lie His Tyr Gin Gin Asn Gin Ala 25 30 Ser cys Gly Lys Arg Ala lie lie Leu Glu Thr Arg Gin His Arg Leu 40 45 Phe Cys Ala Asp Pro Lys Glu Gin Trp Val Lys Asp Ala Met Gin His 50 55 60 Leu Asp Arg Gin Ala Ala Ala Leu Thr Arg Asn Gly Gly Thr Phe Glu 65 70 75 80 Lys Gin lie Gly Glu val Lys Pro Arg Thr Thr Pro Ala Ala Gly Gly 85 90 95 Met Asp Glu Ser val val Leu Glu Pro Glu Ala Thr Gly Glu Ser Ser 100 105 110 Ser Leu Glu Pro Thr Pro Ser Ser Gin Glu Ala Gin Arg Ala Leu Gly 115 120 125 Thr Ser Pro Glu Leu Pro Thr Gly Val Thr Gly Ser Ser Gly Thr Arg 130 135 140 Leu Pro Pro Thr Pro Lys Ala Gin Asp Gly Gly Pro val Gly Thr Glu 145 150 155 160 Leu Phe Arg val Pro Pro Val Ser Thr Ala Ala Thr Trp Gin Ser Ser 165 170 175 Ala Pro His Gin Pro Gly Pro Ser Leu Trp Ala Glu Ala Lys Thr Ser 180 185 190 Glu Ala Pro ser Thr Gin Asp Pro Ser Thr Gin Ala Ser Thr Ala Ser 195 200 205 Ser Pro Ala Pro Glu Glu Asn Ala Pro Ser Glu Gly Gin Arg val Trp 210 215 220 Gly Gin Gly Gin Ser Pro Arg Pro Glu Asn ser Leu Glu Arg Glu Glu 225 230 235 240 Met Gly Pro val Pro Ala His Thr Asp Ala Phe Gin Asp Trp Gly Pro 245 250 255 Gly ser Met Ala His val Ser val val Pro Val Ser Ser Glu Gly Thr 260 265 270 Page 10 PBD00038_Seq . ST25. txt Pro Ser Arg Glu Pro val Ala Ser Gly Ser Trp Thr Pro Lys Ala Glu 275 280 285 Glu Pro lie His Ala Thr Met Asp Pro Gin Arg Leu Gly Val Leu lie 290 295 300 Thr Pro al Pro Asp Ala Gin Ala Ala Thr Arg Arg Gin Ala val Gly 305 310 315 320 Leu Leu Ala Phe Leu Gly Leu Leu Phe Cys Leu Gly val Ala Met Phe 325 330 335 Thr Tyr Gin Leu Gin Gly Cys Pro Arg Met Ala Gly Glu Met 345 350 Ala Glu Gly Leu Arg Tyr lie Pro Arg ser cys Gly Ser Asn Ser Tyr 355 360 365 val Leu al Pro val 370 <210> 21 <211> 76 <212> PRT <213> Homo sapiens <400> 21 Gin Pro val Gly lie Asn Thr Ser Thr Thr Cys Cys Tyr Arg Phe lie 1 5 10 15 Asn Lys Lys lie Pro Lys Gin Arg Leu Glu Ser Tyr Arg Arg Thr Thr 25 30 Ser Ser His Cys Pro Arg Glu Ala val lie Phe Lys Thr Lys Leu Asp 40 45 Lys Glu lie Cys Ala Asp Pro Thr Gin Lys Trp val Gin Asp Phe Met 50 55 60 Lys His Leu Asp Lys Lys Thr Gin Thr Pro Lys Leu 65 70 75 <210> 22 <211> 33 <212> PRT Page 11 PBD00038_Seq . ST25. txt <213> Homo sapiens <400> 22 Gin Pro Leu Pro Asp Cys Cys Arg Gin Lys Thr Cys Ser Cys Arg Leu 1 5 10 15 Tyr Glu Leu Leu His Gly Ala Gly Asn His Ala Ala Gly lie Leu Thr 25 30 Leu <210> 23 <211> 11 <212> PRT <213> Homo sapiens <400> 23 Arg pro Lys Pro Gin Gin Phe Phe Gly Leu Met 1 5 10 Page 12 PBD00038_Seq . ST25. txt SEQUENCE LISTING <110> Probiodrug AG <120> Novel Inhibitors of Glutaminyl Cyclase <130> PBD 00038/WO <140> PCT/EP 2005/001153 <141> 2005-02-04 <150> US 60/542,133 <151> 2004-02-05 <150> US 60/634, 364 <151> 2004-12-08 <150> US 10/838,993 <151> 2004-05-05 <160> 23 <170> Patentln version 3.1 <210> 1 <211> 42 <212> PRT <213> Homo sapiens <400> 1 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu Val His His Gin Lys 1 5 10 15 Leu val Phe Phe Ala Glu Asp val Gly Ser Asn Lys Gly Ala lie lie 25 30 Page 1 PBD00038_Seq . ST25. txt Gly Leu Met Val Gly Gly val val lie Ala 40 <210> 2 <211> 40 <212> PRT <213> Homo sapiens <400> 2 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu val His His Gin Lys 1 5 10 15 Leu val Phe Phe Ala Glu Asp val Gly Ser Asn Lys Gly Ala lie lie 25 30 Gly Leu Met Val Gly Gly Val val 40 <210> 3 <211> 40 <212> PRT <213> Homo sapiens <400> 3 Glu Phe Arg His Asp Ser Gly Tyr Glu val His His Gin Lys Leu Val 1 5 10 15 Phe Phe Ala Glu Asp val Gly Ser Asn Lys Gly Ala lie lie Gly Leu 25 30 Met val Gly Gly val Val lie Ala 40 <210> 4 <211> 38 <212> PRT <213> Homo sapiens <400> 4 Glu Phe Arg His Asp Ser Gly Tyr Glu val His His Gin Lys Leu val 1 5 10 15 Page 2 PBD00038_Seq . ST25. txt Phe Phe Ala Glu Asp val Gly Ser Asn Lys Gly Ala lie He Gly Leu 25 30 Met val Gly Gly val val <210> 5 <211> 11 <212> PRT <213> Artificial sequence <220> <223> Synthetic peptide <220> <221> MOD_RES <222> (ID - - (ID <223> AMIDATION <400> 5 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu 1 5 10 <210> 6 <211> 9 <212> PRT <213> Artificial sequence <220> <223> synthetic peptide <220> <221> MOD_RES <222> (9).. (9) <223> AMIDATION <400> 6 Glu Phe Arg His Asp Ser Gly Tyr Glu 1 5 Page 3 PBD00038_Seq . ST25. txt <210> 7 <211> 21 <212> PRT <213> Artificial sequence <220> <223> synthetic peptide <220> <221> MOD_RES <222> (21).. (21) <223> AMIDATION <400> 7 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu val His His Gin Lys 1 5 10 15 Leu val Phe Phe Ala <210> 8 <211> 19 <212> PRT <213> Artificial sequence <220> <223> synthetic peptide <220> <221> OD_RES <222> (19) .. (19) <223> AMIDATION <400> 8 Glu Phe Arg His Asp Ser Gly Tyr Glu val His His Gin Lys Leu Val 1 5 10 15 Phe Phe Ala Page 4 PBD00038_Seq . ST25. txt <210> 9 <211> 38 <212> PRT <213> Artificial sequence <220> <223> Synthetic peptide <400> 9 Gin Phe Arg His Asp Ser Gly Tyr Glu val His His Gin Lys Leu Val 1 5 10 15 Phe Phe Ala Glu Asp val Gly Ser Asn Lys Gly Ala lie lie Gly Leu 25 30 Met val Gly Gly val val <210> 10 <211> 19 <212> PRT <213> Artificial sequence <220> <223> synthetic peptide <220> <221> MOD_RES <222> (19) .. (19) <223> AMIDATION <400> 10 Gin Phe Arg His Asp ser Gly Tyr Glu val His His Gin Lys Leu 1 5 10 15 Phe Phe Ala <210> 11 <211> 11 <212> PRT Page 5 PBD00038_Seq . ST25. txt <213> Artificial sequence <220> <223> Synthetic peptide <220> <221> MOD_RES <222> (11) .. (11) <223> AMIDATION <400> 11 Asp Ala Gin Phe Arg His Asp Ser Gly Tyr Glu 1 5 10 <210> 12 <211> 9 <212> PRT <213> Artificial sequence <220> <223> Synthetic peptide <220> <221> OD_RES <222> (9) .. (9) <223> AMIDATION <400> 12 Gin Phe Arg His Asp Ser Gly Tyr Glu 1 5 <210> 13 <211> 17 <212> PRT <213> Homo sapiens <220> <221> MOD_RES Page 6 PBD00038_Seq . ST25. txt <222> (17).. (17) <223> AMIDATION <400> 13 Gin Gly Pro Trp Leu Glu Glu Glu Glu Glu Ala Tyr Gly Trp Met Asp 1 5 10 . 15 Phe <210> 14 <211> 13 <212> PRT <213> Homo sapiens <400> 14 Gin Leu Tyr Glu Asn Lys Pro Arg Arg Pro Tyr lie Leu 1 5 10 <210> 15 <211> 10 <212> PRT <213> Homo sapiens <220> <221> MOD_RES <222> (10) .. (10) <223> AMIDATION <400> 15 Gin His Trp Ser Tyr Gly Leu Arg Pro Gly 1 5 10 <210> 16 <211> 97 <212> PRT <213> Homo sapiens Page 7 PBD00038_Seq . ST25. txt <400> 16 Gin Pro Lys val Pro Glu Trp val Asn Thr Pro ser Thr Cys Cys Leu 1 5 10 15 Lys Tyr Tyr Glu Lys val Leu Pro Arg Arg Leu val Val Gly Tyr Arg 25 30 Lys Ala Leu Asn Cys His Leu Pro Ala lie lie Phe val Thr Lys Arg 40 45 Asn Arg Glu val Cys Thr Asn Pro Asn Asp Asp Trp val Gin Glu Tyr 50 55 60 lie Lys Asp Pro Asn Leu Pro Leu Leu Pro Thr Arg Asn Leu ser Thr 65 70 75 80 val Lys lie lie Thr Ala Lys Asn Gly Gin Pro Gin Leu Leu Asn Ser 85 90 95 Gin <210> 17 <211> 76 <212> PRT <213> Homo sapiens <400> 17 Gin Pro Asp Ser val Ser lie Pro lie Thr Cys Cys Phe Asn val lie 1 5 10 15 Asn Arg Lys lie Pro lie Gin Arg Leu Glu ser Tyr Thr Arg lie Thr 25 30 Asn lie Gin Cys Pro Lys Glu Ala val lie Phe Lys Thr Lys Arg Gly 40 45 Lys Glu Val Cys Ala Asp Pro Lys Glu Arg Trp val Arg Asp ser Met 50 55 60 Lys His Leu Asp Gin lie Phe Gin Asn Leu Lys Pro 65 70 75 <210> 18 <211> 76 <212> PRT Page 8 PBD00038_seq . ST25. txt <213> Homo sapiens <400> 18 Gin Pro Asp Ala lie Asn Ala Pro val Thr Cys Cys Tyr Asn Phe Thr 1 5 10 15 Asn Arg Lys lie Ser Val Gin Arg Leu Ala Ser Tyr Arg Arg lie Thr 25 30 Ser Ser Lys cys Pro Lys Glu Ala Val lie Phe Lys Thr lie Val Ala 40 45 Lys Glu lie Cys Ala Asp Pro Lys Gin Lys Trp val Gin Asp Ser Met 50 55 60 Asp His Leu Asp Lys Gin Thr Gin Thr Pro Lys Thr 65 70 75 <210> 19 <211> 68 <212> PRT <213> Homo sapiens <400> 19 Gin val Gly Thr Asn Lys Glu Leu Cys Cys Leu Val Tyr Thr Ser Trp 1 5 10 15 Gin lie Pro Gin Lys Phe lie val Asp Tyr Ser Glu Thr Ser Pro Gin 25 30 Cys Pro Lys Pro Gly val lie Leu Leu Thr Lys Arg Gly Arg Gin lie 40 45 Cys Ala Asp Pro Asn Lys Lys Trp Val Gin Lys Tyr lie Ser Asp Leu 50 55 60 Lys Leu Asn Ala 65 <210> 20 <211> 373 <212> PRT <213> Homo sapiens Page 9 PBD00038_Seq . ST25. txt <400> 20 Gin His His Gly Val Thr Lys Cys Asn lie Thr Cys Ser Lys Met Thr 1 5 10 15 Ser Lys lie Pro Val Ala Leu Leu lie His Tyr Gin Gin Asn Gin Ala 25 30 Ser cys Gly Lys Arg Ala lie lie Leu Glu Thr Arg Gin His Arg Leu 40 45 Phe Cys Ala Asp Pro Lys Glu Gin Trp val Lys Asp Ala Met Gin His 50 55 60 Leu Asp Arg Gin Ala Ala Ala Leu Thr Arg Asn Gly Gly Thr Phe Glu 65 70 75 80 Lys Gin lie Gly Glu val Lys Pro Arg Thr Thr Pro Ala Ala Gly Gly 85 90 95 Met Asp Glu ser val Val Leu Glu Pro Glu Ala Thr Gly Glu Ser Ser 100 105 110 Ser Leu Glu Pro Thr Pro Ser ser Gin Glu Ala Gin Arg Ala Leu Gly 115 120 125 Thr Ser Pro Glu Leu Pro Thr Gly val Thr Gly Ser Ser Gly Thr Arg 130 135 140 Leu Pro Pro Thr pro Lys Ala Gin Asp Gly Gly Pro val Gly Thr Glu 145 150 155 160 Leu Phe Arg Val Pro Pro val ser Thr Ala Ala Thr Trp Gin Ser Ser 165 170 175 Ala Pro His Gin Pro Gly Pro Ser Leu Trp Ala Glu Ala Lys Thr Ser 180 185 190 Glu Ala Pro Ser Thr Gin Asp Pro Ser Thr Gin Ala Ser Thr Ala Ser 195 200 205 Ser Pro Ala Pro Glu Glu Asn Ala Pro Ser Glu Gly Gin Arg Val Trp 210 215 220 Gly Gin Gly Gin Ser Pro Arg Pro Glu Asn Ser Leu Glu Arg Glu Glu 225 230 235 240 Met Gly Pro Val Pro Ala His Thr Asp Ala Phe Gin Asp Trp Gly Pro 245 250 255 Gly Ser Met Ala His val Ser val val Pro val Ser Ser Glu Gly Thr 260 265 270 Page 10 PBD00038_Seq . ST25. txt Pro ser Arg Glu Pro val Ala Ser Gly Ser Trp Thr Pro Lys Ala Glu 275 280 285 Glu Pro lie His Ala Thr Met Asp Pro Gin Arg Leu Gly val Leu lie 290 295 300 Thr Pro val Pro Asp Ala Gin Ala Ala Thr Arg Arg Gin Ala val Gly 305 310 315 320 Leu Leu Ala Phe Leu Gly Leu Leu Phe Cys Leu Gly Val Ala Met Phe 325 330 335 Thr Tyr Gin Ser Leu Gin Gly Cys Pro Arg Lys Met Ala Gly Glu Met 340 345 350 Ala Glu Gly Leu Arg Tyr lie Pro Arg ser Cys Gly Ser Asn Ser Tyr 355 360 365 val Leu val Pro Val 370 <210> 21 <211> 76 <212> PRT <213> Homo sapiens <400> 21 Gin Pro val Gly lie Asn Thr Ser Thr Thr Cys Cys Tyr Arg Phe lie 1 5 10 15 Asn Lys Lys lie Pro Lys Gin Arg Leu Glu Ser Tyr Arg Arg Thr Thr 25 30 Ser Ser His Cys Pro Arg Glu Ala Val lie Phe Lys Thr Lys Leu Asp 40 45 Lys Glu lie Cys Ala Asp Pro Thr Gin Lys Trp val Gin Asp Phe Met 50 55 60 Lys His Leu Asp Lys Lys Thr Gin Thr Pro Lys Leu 65 70 75 <210> 22 <211> 33 <212> PRT Page 11 PBD00038_Seq . ST25. txt <213> Homo sapiens <400> 22 Gin Pro Leu Pro Asp Cys Cys Arg Gin Lys Thr Cys Ser Cys Arg Leu 1 5 10 15 Tyr Glu Leu Leu His Gly Ala Gly Asn His Ala Ala Gly lie Leu Thr 25 30 Leu <210> 23 <211> 11 <212> PRT <213> Homo sapiens <400> 23 Arg Pro Lys Pro Gin Gin Phe Phe Gly Leu Met 1 5 10 Page 12

Claims (27)

1. 08 176765/3 Claims 1. A compound of the formula 1 including pharmaceutically acceptable salts thereof, including all stereoisomers and polymorphs: I N-A-B ^ formula 1 wherein: A is an unbranched C3 alkyl chain; and B is a group selected from (VI) or (VII): (VI) (VII) wherein: D represents substituted phenyl, wherein substitution means -oxyalkyi, - thioalkyl, halogenyl; or D represents dihydrobenzodioxine, benzodioxole, benzodithiole, dihydrobenzodithiine, benzooxathiole or dihydrobenzooxathiine; X represents S, N-CN; Y is S; R17 and R 8 are both H; one of R17 and R18 is H and the other is methyl; or R17 and R18 can be connected to form a carbocycle with up to 6 ring atoms; and wherein the term alkyl means methyl, ethyl, propyl or butyl, with the proviso (f) that the compounds 109 176765/3 are excluded from formula 1 ; or a compound of the formula 1a including pharmaceutically acceptable salts thereof, including all stereoisomers and polymorphs: 110 176765/3 wherein the group R represents methyl, ferf-butyl, benzyl, phenyl, 4-ethyl-phenyl, 4-(methoxycarbonyl)-phenyl, 4-(methylcarbonyl)-phenyl, bicyclo[2.2.1]hept-5-en-2-yl, 2-(methoxycarbonyl)-phenyl, 4-(oxazol-5-yl)-phenyl, 4-(pyrazol-1-yl)-phenyl, 4-isopropyl-phenyl, 4-(piperidine-1-sulfonyl)-phenyl, 4-(morpholin-4-yl)-phenyl, 4-cyano-phenyl, 2,3-dihydro-benzo[1 ,4]-dioxin-6-yl, benzo[1 ,3]dioxol-5-yl, 4-(benzyloxy)-phenyl, 4-(methoxy)-benzyl, 4-methyl-phenyl, naphthalen-1-yl, 4-nitro-phenyl, n-butyl, cyclooctyl, furan-2-yl-methyl, tetrahydrofuran-2-yl-methyl, benzo[1,3]dioxol-5-ylmethyl, 2-(morpholin-4-yl)-ethyl, 4-(dimethylamino)-phenyl, 4-(trifluoromethoxy)-phenyl, benzoyl or pyridin-4-yl; with the proviso (e) that the following compounds are excluded: or a compound of the formula 1b including pharmaceutically acceptable salts thereof, including all stereoisomers and polymorphs: wherein the group R2 represents methyl, te/f-butyl, benzyl, phenyl, 4-ethyl-phenyl, 4-(trifluoromethyl)-phenyl, 4-(methylcarbonyl)-phenyl, 2,3-dihydro-benzo[1 ,4]-dioxin-6-yl, benzo[d][1 ,3]dioxol-6-yl, 4-(benzyloxy)-phenyl, 4-methyl-phenyl, naphthalen-1-yl, 4-nitro-phenyl, n-butyl, cyclooctyl, furan-2-yl-methyl, tetrahydrofuran-2-yl-methyl, 111 176765/3 benzo[d][1 ,3]dioxol-6-ylmethyl, 2-(morpholin-4-yl)-ethyl, 4-(dimethylamino)-phenyl, or trityl.
2. The compound of formula 1 according to claim 1, wherein B is group (VI).
3. The compound of formula 1 according to claim 2, wherein X represents S.
4. The compound of formula 1 according to claim 1 , wherein B is group (VII).
5. The compound of formula 1 according to claim 4, wherein Y represents S.
6. The compound of formula 1 according to either claim 4 or claim 5, wherein R17 and R18 are both H.
7. The compound of formula 1 according to either claim 4 or claim 5, wherein one of R 7 and R18 is H and the other is methyl.
8. The compound of formula 1 according to either claim 4 or claim 5, wherein R17 and R18 are connected to form a carbocycle with up to 6 ring atoms.
9. The compound of formula 1 according to claim 1 , wherein X represents S and the group D represents 4-fluoro-phenyl, 4-chloro-phenyl, 4-(trifluoromethyl)-phenyl, 4-(methoxy)-phenyl, 3,4-(dimethoxy)-phenyl, 2,4-(dimethoxy)-phenyl, 3,5-(dimethoxy)-phenyl, 3,4,5-(trimethyoxy)-phenyl, 3-(methoxy)-phenyl, 4-(ethoxy)-phenyl, 4-iodo-phenyl, 4-bromo-phenyl or 4-(methylsulfanyl)-phenyl.
10. The compound of the formula 1 according to claim 9, wherein the group D represents 3,4-(dimethoxy)-phenyl.
11. The compound of formula 1 according to claim 1 , wherein X represents N-CN and the group D represents 4-fluoro-phenyl, 4-(methoxy)-phenyl, 3,4-(dimethoxy)-phenyl, 2,4-(dimethoxy)-phenyl, 2,3,4-(trimethoxy)-phenyl, 3,5-(dimethoxy)-phenyl, 3- 112 176765/3 (methoxy)-phenyl, 4-(ethoxy)-phenyl, 4-iodo-phenyl, 4-bromo-phenyl, or 4-(methyltsulfanyl)-phenyl;
12. The compound of the formula 1 according to claim 1 , wherein said compound of formula 1 corresponnds to formula 1g wherein: the group R7 represents 3,4-(dimethoxy)-phenyl and the groups R8 and R9 are both H; or the group R7 represents 4-(chloro)-phenyl and the groups R8 and R9 together with the carbon atom to which they are attached form a cyclobutyl or cyciopentyl ring; or the group R7 represents 4-(methoxy)-phenyl and the groups R8 and R9 together with the carbon atom to which they are attached form a cyclopropyl or cyclohexyl ring.
13. The compound of the formula 1 g according to claim 12, wherein the group R7 represents 3,4-(dimethoxy)-phenyl and the groups R8 and R9 are both H.
14. The compound of the formula 1g according to claim 12, wherein the groups R8 and R9 together with the carbon atom to which they are attached form a cyclopropyl ring and the group R7 represents 3,4-(dimethoxy)-phenyl.
15. The compound according to any one of the claims 1 to 14 for use as a pharmaceutical.
16. A pharmaceutical composition comprising at least one compound according to any one of the claims 1 to 14 optionally in combination with a therapeutically acceptable carrier and/or excipient. 113 176765/3
17. The pharmaceutical composition according to claim 16 for parenteral, enteral or oral administration.
18. The pharmaceutical composition of claim 16 or 17, which comprises additionally at least one compound, selected from the group consisting of PEP-inhibitors, LiCI, inhibitors of dipeptidyl aminopeptidases, preferably inhibitors of DP IV or DP IV-like enzymes, NPY-receptor ligands, NPY agonists, ACE inhibitors, PIMT enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of neutral endopeptidase, PDE-4 inhibitors, MAO inhibitors, TNFalpha inhibitors, amyloid protein or amyloid peptide deposition inhibitors, sigma-1 receptor inhibitors and histamine H3 antagonists.
19. The pharmaceutical composition according to claim 18, wherein said inhibitor of DP IV/DP IV-like enzymes is selected from the group consisting of L-threo-isoleucyl pyrrolidide, L-allo-isoleucyl thiazolidide, L-allo-isoleucyl pyrrolidide; and salts thereof or valine pyrrolidide, BMS-477118, CP-867534-01 , LAF-237, PHX-1004, SSR-162369, SYR-322, TSL-225, FE-999011 GW-229A, 815541, K-579, MK-431, PT-100 or one of and 114 176765/3
20. The pharmaceutical composition according to claim 18, wherein said NPY antagonist is selected from 3a,4,5,9b-tetrahydro-1h-benz[e]indol-2-yl amine, BIBP3226 and, (R)-N2-(diphenylacetyl)-(R)-N-[1-(4-hydroxy- phenyl) ethyl] arginine amide.
21. The pharmaceutical composition according to claim 18, wherein said PEP-inhibitor is selected from the group consisting of chemical derivatives of proline or small peptides containing terminal prolines, e.g. benzyloxycarbonyl-prolyl-prolinal, N-terminal substituted L-proline or L-prolylpyrrolidine, substituted N-benzyloxycarbonyl (Z) dipeptides containing prolinal at the carboxy terminus, substituted thioprolines, substituted thiazolidines, substituted oxopyrrolidines, carboxy terminal modified prolines including fluorinated ketone derivatives, chloromethyl ketone derivatives of acyl-proline or acylpeptide-proline (Z-Gly-Pro-ChbCI) and 2-acylpyrrolidine derivatives.
22. The pharmaceutical composition according to claim 18, wherein said PEP-inhibitor is selected from the group consisting of Fmoc-Ala-Pyrr-CN, Z-321 , ONO-1603, JTP-4819 and S-17092.
23. The pharmaceutical composition according to claim 18, wherein said PEP-inhibitor is 115 176765/3
24. The pharmaceutical composition according to claim 18, wherein said ACE-inhibitor is SDZ ENA 713 (rivastigmine (+)-(S)-N-ethyl-3-[(1-dimethylamino)ethyl]-N-methylphenylcarbamate hydrogen tartrate.
25. The pharmaceutical composition according to claim 18, wherein said PDE-4 inhibitor is selectede from the group consisting of Rolipram, CC-002, L-826141 , Sch-351591 (D-4396), OS-0217, IBFB-130011 , IBFB-150007, IBFB-130020, IBFB-140301 , IC-485, VMX-554, VMX-565, MEM-1414, MEM-1018, MEM-1091 , MEM-1145, CI-1044, BHN, ZK-117137 and SB-207499 or analogs thereof.
26. The pharmaceutical composition according to claim 18, wherein said PIMT enhancer is a 10-aminoaliphatyl-dibenz[b, f] oxepine of the general formula wherein alk is a divalent aliphatic radical, R is an amino group that is unsubstituted or mono- or di-substituted by monovalent aliphatic and/or araliphatic radicals or disubstituted by divalent aliphatic radicals, and Ri, R2, R3 and R4 are each, independently of the others, hydrogen, lower alkyl, lower alkoxy, halogen or trifluoromethyl.
27. The pharmaceutical composition according to claim 18, wherein said gamma secretase inhibitor is 29. The pharmaceutical composition according to claim 18, wherein said MAO inhibitor is ladostigil of the formula 30. The pharmaceutical composition according to claim 18, wherein said histamine H3 antagonist is a compound selected from the group consisting of A-331440, A-349821 , 3874-H1 , UCL-2173, UCL-1470, DWP-302, GSK-189254A, GSK-207040A, cipralisant, GT-2203, 1S,2S)-2-(2-Aminoethyl)-1-(1 H-imidazol-4-yl)cyclopropane, JNJ-5207852, NNC-0038-0000-1049, dual H1/H3, Sch-79687 or one of 1 17 176765/3 31. Use of a compound according to any of claims 1 to 14 or a pharmaceutical composition according to any of the claims 16 to 30 for the preparation of a medicament for the treatment of neuronal disorders, especially Alzheimer's disease, Down Syndrome, Parkinson disease, Chorea Huntington, pathogenic psychotic conditions, schizophrenia, impaired food intake, sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance, impaired regulation, body fluids, hypertension, fever, sleep dysregulation, anorexia, anxiety related disorders including depression, seizures including epilepsy, drug withdrawal and alcoholism, neurodegenerative disorders including cognitive dysfunction and dementia. 32. The use according to claim 31 for the treatment of a neuronal disease selected from the group consisting of Alzheimer's disease, Down Syndrome, Parkinson disease and Chorea Huntington. 33. The use according to claims 31 or 32, wherein said inhibitor inhibits glutaminyl cyclase with a Ki of 10 μΜ or less. 118 176765/1 34. The use according to any one of claims 31 to 33, wherein said inhibitor inhibits glutaminyl cyclase with a K\ of 1 μΜ or less. 35. The use according to any one of claims 31 to 34, wherein said inhibitor inhibits glutaminyl cyclase with a Kj of 0.1 μΜ or less. 36. The use according to any one of claims 31 to 35, wherein said inhibitor inhibits glutaminyl cyclase with a K, of 0.01 μΜ or less. 37. The use according to any one of claims 31 to 36, wherein said glutaminyl cyclase inhibitor has a molecular weight of 1000 g/mole or less. 38. The use according to any one of claims 31 to 37, wherein said glutaminyl cyclase inhibitor has a molecular weight of 500 g/mole or less. 39. The use according to any one of claims 31 to 38, wherein said glutaminyl cyclase inhibitor has a molecular weight of 400 g/mole or less. 40. The use according to any one of claims 31 to 39, wherein said glutaminyl cyclase inhibitor has a molecular weight of 350 g/mole or less. 41. The use according to any one of claims 31 to 40, wherein said glutaminyl cyclase inhibitor is a competitive inhibitor. 42. The use according to any one of claims 31 to 41 , wherein said glutaminyl cyclase inhibitor is a competitive reversible inhibitor. For the Applicants, REINHOLD COHN PARTNERS
IL176765A 2004-02-05 2006-07-10 Inhibitors of glutaminyl cyclase IL176765A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US54213304P 2004-02-05 2004-02-05
US10/838,993 US7371871B2 (en) 2003-05-05 2004-05-05 Inhibitors of glutaminyl cyclase
US63436404P 2004-12-08 2004-12-08
PCT/EP2005/001153 WO2005075436A2 (en) 2004-02-05 2005-02-04 Novel inhibitors of glutaminyl cyclase

Publications (2)

Publication Number Publication Date
IL176765A0 IL176765A0 (en) 2006-10-31
IL176765A true IL176765A (en) 2011-08-31

Family

ID=42121404

Family Applications (1)

Application Number Title Priority Date Filing Date
IL176765A IL176765A (en) 2004-02-05 2006-07-10 Inhibitors of glutaminyl cyclase

Country Status (5)

Country Link
AT (1) ATE541835T1 (en)
DK (1) DK1713780T3 (en)
EA (1) EA013790B1 (en)
HK (1) HK1100943A1 (en)
IL (1) IL176765A (en)

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS605593B2 (en) * 1981-03-20 1985-02-12 北興化学工業株式会社 Imidazole derivatives and agricultural and horticultural fungicides
EP0912551A1 (en) * 1996-05-14 1999-05-06 Novo Nordisk A/S Somatostatin agonists and antagonists
WO1999020599A1 (en) * 1997-10-21 1999-04-29 Cambridge Neuroscience, Inc. Pharmaceutically active compounds and methods of use
IL140197A0 (en) * 1998-06-12 2002-02-10 Sod Conseils Rech Applic β-CARBOLINE COMPOUNDS
CN100439332C (en) * 2000-08-21 2008-12-03 株式会社太平洋 Novel thiourea derivatives and the pharmaceutical compositions containing thd same
SE0100566D0 (en) * 2001-02-20 2001-02-20 Astrazeneca Ab Compounds
EP1620082B9 (en) * 2003-05-05 2010-08-25 Probiodrug AG Medical use of inhibitors of glutaminyl and glutamate cyclases for treating alzheimer's disease and down syndrome
DE602004026289D1 (en) * 2003-05-05 2010-05-12 Probiodrug Ag Glutaminyl INHIBITORS

Also Published As

Publication number Publication date
ATE541835T1 (en) 2012-02-15
HK1100943A1 (en) 2007-10-05
EA200601411A1 (en) 2007-02-27
DK1713780T3 (en) 2012-05-07
EA013790B1 (en) 2010-06-30
IL176765A0 (en) 2006-10-31

Similar Documents

Publication Publication Date Title
US7304086B2 (en) Inhibitors of glutaminyl cyclase
US7371871B2 (en) Inhibitors of glutaminyl cyclase
US7732162B2 (en) Inhibitors of glutaminyl cyclase for treating neurodegenerative diseases
CA2735795C (en) Novel inhibitors
KR20190136005A (en) Novel inhibitors
DK2686313T3 (en) Benzimidazole derivatives as inhibitors of glutaminyl cyclase
NZ546887A (en) Combinations useful for the treatment of neuronal disorders
JP2010511013A (en) Novel inhibitors of glutaminyl cyclase
IL176765A (en) Inhibitors of glutaminyl cyclase
NZ548596A (en) Novel inhibitors of glutaminyl cyclase and their use in treating neuronal diseases
EVANS Schilling et al.(43) Pub. Date: Sep. 29, 2005
MXPA06008868A (en) Novel inhibitors of glutaminyl cyclase
CENTER Schilling et al.(43) Pub. Date: Nov. 11, 2004
ES2348500T3 (en) MEDICAL USE OF GLUTAMINYL AND GLUTAMATE CYCLASE INHIBITORS FOR THE TREATMENT OF ALZHEIMER&#39;S DISEASE AND DOWN SYNDROME.

Legal Events

Date Code Title Description
FF Patent granted
KB Patent renewed
KB Patent renewed
KB Patent renewed