OA16479A - Pyrazole compounds as CRTH2 antagonists. - Google Patents

Pyrazole compounds as CRTH2 antagonists. Download PDF

Info

Publication number
OA16479A
OA16479A OA1201300282 OA16479A OA 16479 A OA16479 A OA 16479A OA 1201300282 OA1201300282 OA 1201300282 OA 16479 A OA16479 A OA 16479A
Authority
OA
OAPI
Prior art keywords
formula
alkyl
pyrazole compounds
compounds
alkoxy
Prior art date
Application number
OA1201300282
Inventor
Ralf Anderskewitz
Domnic Martyres
Thorsten Oost
Wolfgang Rist
Peter Seither
Original Assignee
Boehringer Ingelheim International Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boehringer Ingelheim International Gmbh filed Critical Boehringer Ingelheim International Gmbh
Publication of OA16479A publication Critical patent/OA16479A/en

Links

Abstract

The present invention relates to pyrazole compounds of formula (1a) or (1b) and pharmaceutically acceptable salts thereof,

Description

PYR, \ZOLE COMPOUNDS AS CRTH2 ANTAGONISTS
The présent invention relates to pyrazole compounds of formula (la) or (Ib) and pharmaceutically acceptable salts thereof having CRTH2 antagonistic activity,
wherein Ra, Rb, Rc, Rd, Y1, Y2, Y3, Y4, Y5, Z, R1, R2, n and R3 hâve one of the meanings as indicated in the spécification and claims, to their use as médicaments, to pharmaceutical formulations containing said compounds and to pharmaceutical formulations said compounds in combination with one or more active substances,
BACKGROUND OF THE INVENT ION
Prostaglandin D2 (PGD2) is an eicosanoid generated by the metabolism of arachidonic acids upon stimulation of inflammatory cells with allergens, inflammatory stimuli or by tissue damage, PGD2 is primarily released by mast cells with Th2 cells, dendritic cells, and macrophages being secondary sources. PGD2 is the major arachidonic acid métabolite produced by mast cells upon allergen challenge (Lewis et al,, J, Immunol. 1982, 129:16271631) and has been detected in high concentrations in the airways of asthmatic patients (Murray et al, N. Engl. J. Med., 1986, 315:800-804; Liu et al., Am. Rev. Respir Dis., 1990, 142 126-132; Zehretal., Chest, 1989, 95:1059-63; Wenzel étal., J. Allergy. Clin. Immunol., j
-116479
2/64
1991, 87540-548). PGD2 production is also increased in patients with systemic mastocytosis (Roberts N. Engl. J. Med. 1980, 303, 1400-1404; Butterfield étal., Int Arch Allergy Immunol, 2008,147:338-343) allergie rhinitis (Naclerio et al., Am. Rev. Respir. Dis., 1983, 128:597-602; Brown et al., Arch Otolaryngol Head Neck Surg, 1987,113:179-183; Lebel étal., J. Allergy s Clin. Immunol., 1988, 82:869-877), urticaria (Heavy et al., J. Allergy. Clin. Immunol., 1986, 78:458-461), chronic rhinosinusitis (Yoshimura étal., Allergol. Int., 2008, 57:429-436), chronic obstructive pulmonary disease (Csanky et al., Electrophoresis, 2009, 30:1228-1234) and during anaphylaxis (Ono et al., Clin. Exp. Allergy, 2009, 39:72-80).
io Instillation of PGD2 into airways can provoke features of asthmatic response including bronchoconstriction (Hardy et al., 1984, N Engl J. Med 311:209-213; Sampson et al 1997, Thorax 52: 513-518) and eosinophil accumulation (Emery et al., 1989, J. Applied Physiol 67: 959-962). The potential of PGD2 to trigger inflammatory responses has been confirmed by the overexpression of human PGD2 synthase in mice resulting in elevated eosinophil iung inflammation and Th2 cytokine production in response to allergen (Fujitani et al, 2002 J. Immunol. 168:443-449).
PGD2 is an agonist of two 7-transmembrane type G protein-coupled receptors, the PGD2 receptor DP1 (Boie et al., J Biol Chem, 1995, 270:18910-6) and the recently identified
CRTH2 (chemoattractant receptor-homologous molécule expressed on Th2 cells) receptor (also referred to as DP2 receptor) (Nagata et al., J. Immunol., 1999, 162:1278-86).
CRTH2 is expressed on Th2 cells, eosinophils, basophils and mast cells (Nagata et al., FEBS Lett, 1999, 459: 195-199; Nagata et al., J Immunol, 1999, 162: 1278-1286; Cosmi et 25 al., Eur J Immunol, 2000, 30:2972-2979; Boehme et al., Int Immunol, 2009, 21: 621-32).
Using sélective CRTH2 agonists like 13,14 dihydro-15-keto-PGD2 (DK-PGD2) and 15Rmethyl-PGD2, it has been shown that CRTH2 activation initiâtes cellular processes that lead to the recruitment and activation of inflammatory cells (Spik et al., J. Immunol., 2005;174:3703-8; Shiraishi, J. Pharmacol. Exp. Ther., 2005, 312:954-60; Monneret et al., J.
Pharmacol. Exp. Ther., 2003, 304:349-355). Using CRTH2 sélective antagonists it has been shown that inflammatory responses and pathophysiological changes in animal models of diseases like asthma, allergie rhinitis, atopie dermatitis and COPD can be diminished (Uller et al., Respir Res. 2007, 8:16; Lukacs et al., Am. J. Physiol. Lung Cell Mol. Physiol. 2008,
295:L767-79; Stearns, Bioorg. Med. Chem. Lett. 2009,19:4647-51; Nomiya, J Immunol,
2008, 180:5680-5688; Boehme étal., Int. Immunol., 2009, 21:1-17; Boehme et al., Int
-216479
3/64
Immunol, 2009, 21:81-93; Takeshita et al., Int Immunol, 2004, 16:947-59; Stebbins étal., J. Pharmacol. Exp. Ther. 2009). Moreover, genetic délétion of CRTH2 in mice diminished inflammatory responses in animal models of allergy (Shiraishi et al., J. Immunol. 2008;180:541-549; Oiwa, Clin Exp Allergy, 2008, 38:1357-66; Satoh étal., J. Immunol., 2006,177:2621-9). In contrast, the sélective DP1 agonist BW245C does not promote inflammatory responses, like migration or activation of Th2 lymphocytes, basophils or eosinophils (Yoshimura-Uchiyama et al., Clin. Exp. Allergy, 2004, 34:1283-90; Xue et al., Immunol, 2005, 175:6531-6; Gervais et al., J. Allergy Clin. Immunol., 2001, 108:982-8). Therefore, agents that antagonize the effects of PGD2 at the CRTH2 receptor should be useful for the treatment of respiratory or gastrointestinal complaints, as well as inflammatory diseases of the joints and allergie diseases of the nasopharynx, eyes and skin.
WO 2004/096777 teaches pyrimidine dérivatives of formula (a) and salts thereof,
wherein R6 is carboxy, carboxamide, nitrile or tetrazolyl, said dérivatives having CRTH2 antagonistic activity and can be used for the prophylaxis and treatment of diseases associated with CRTH2 activity.
WO 2009/042138 daims alkylthio substituted pyrimidine compounds of formula (b),
-316479
4/64 said compounds having CRTH2 antagoniste activity.
WO 2009/042139 claims 2-S-benzyl pyrimidine compounds of formula (c),
said compounds having CRTH2 antagoniste activity.
EP 0 480 659 claims compounds of general formula (d),
Y wherein Z2 inter alia may be carboxyl-C1-C10-alkyl-C= and Y may be substituted benzyl, said compounds being useful for the treatment of hyperuricemia.
WO 2005/040128 claims compounds of general formula (e),
said compounds being useful for the treatment of conditions such as pain, or an inflammatory, immunological, bone, neurodegenerative or rénal disorder.
WO 01/38325 claims compounds of general formula (f),
-416479
5/64 wherein A is an aromatic ring and B is a nitrogen-containing 5-membered hetero ring which may further be substituted, said compounds having hypoglycémie and hypolipidémie activity.
It is an objective of the présent invention to provide further compounds having CRTH2 antagoniste activity.
Preferably the compounds of the présent invention hâve enhanced chemical stability, enhanced pharmacokinetic properties (PK) and/or enhanced activity in a whole cell assay.
DETAILED DESCRIPTION OF THE INVENTION
The présent invention relates to pyrazole compounds of formula (la) or (Ib) and pharmaceutically acceptable salts thereof,
wherein
Ra and Rb are îndependently selected hydrogen, hydroxy, halogen, CrCe-alkyl, CrC6 haloalkyl, CrC6-alkoxy, CrC6-haloalkoxy and C3-Ce-cycloalkyl, or Ra and Rb together 20 with the carbon atom they are bound to form may form a carbonyl group, or Ra and Rb together with the carbon atom they are bound to form a 3- to 8-membered ring, wherein said ring may contain 1 or 2 heteroatoms selected from O, N and S as ring member and wherein the ring members of said ring may optionally be îndependently
-516479
6/64 substituted by hydroxy, halogen, Ci-Ce-alkyl, CrCe-haloalkyl, CrCe-alkoxy, CrC6haloalkoxy and C3-C8-cycloalkyl;
R° and Rd are independently selected hydrogen, hydroxy, halogen, Cf-Ce-alkyi, ΟΊΒ5 haloalkyl, CrCB-alkoxy, Ci-C6-haloalkoxy and C3-C8-cycloalky|, or Rc and Rd together with the carbon atom they are bound to form may form a carbonyl group, or Rc and Rd together with the carbon atom they are bound to form a 3- to 8-membered ring, wherein said ring may contain 1 or 2 heteroatoms selected from O, N and S as ring member and wherein the ring members of said ring may optionally be independently io substituted by hydroxy, halogen, CrCe-alkyl, CrCc-haloalkyl, CrCe-alkoxy, CrC6haloalkoxy and C3-Ce-cycloalkyl;
Y1, Y2, Y3, Y4 and Y5 are independently selected from N and CRy, wherein each Ry is independently selected from H, hydroxy, halogen, cyano, nitro, SFs, C(O)NRfR9, Cr 15 C6-alkyl, hydroxy-CrCB-alkyl, CrCe-alkoxy-CrCe-alkyl, C3- Ca-cycloalkyî, Ο,-Οβhaloalkyl, CpCe-alkoxy, Ο,-Ce-alkoxy-Ci-Ce-alkoxy, CrCe-haloalkoxy, C3-C8cycloalkoxy, CrCs-alkylamino, di-CrCe-alkylamino, CrCB-alkylsulfonyl, phenyl, phenoxy, 5- or 6-membered heterocyclyl and 5- or 6-membered heterocyclyloxy, wherein Rf and R9 are independently from each other selected from H, CrCe-alkyl,
CrCe-haloalkyl, C3-Ca-cycloalkyl, C3-C8-cycloalkenyl and 5- or 6-membered heterocyclyl or Rf and R9 together with the nitrogen atom to which they are bound form a cyclic amine, which may comprise a further heteroatom selected from O, N and S as a ring member;
Z is selected from O, S and NRZ, wherein Rz is H, CrCe-alkyl or benzyl;
R1 and R2 are independently from each other selected from H, halogen, CrCB-alkyl, C2-CBalkenyl, C2-C6-alkynyl, Ci-C6-alkoxy, CrCB-alkylthio, -NRfR9, C3-C8-cycloalkyl, C3-CBcycloalkyl-CrCe-alkyl, C3-C8-cycloalkyl-C2-C6-alkenyl, C3-C8-cycloalkenyl, C3-C830 cycloalkenyl-CrCe-alkyl, C3-C6-cycloalkeny!-C2-C6-alkenyl, phenyl, phenyl-CrC6-alkyl, phenyl-C2-C6-alkenyl, naphthyl, naphthyl-CrCe-alkyl, naphthyl-C2-CB-alkenyl, heterocyclyl, heterocyclyî-Ci-CB-alkyl, and heterocyclyl-C2-C6-alkenyl, wherein the CpCe-alkyl, C2-CB-alkenyl and C2-C6-alkynyl moieties in the aforementioned radicale R1 and R2 are unsubstituted or carry at least one substituent selected from
-616479
7/64 hydroxy, halogen, cyano, nitro, CrC6-alkoxy, CrCe-haloalkoxy, Ci-C6-alkylamino, di
Ci-Ce-alkylamino and Ci-C6-alkylsulfonyl and/or wherein two radicals bound to the same carbon atom of said Ci-C6-alkyl, C2-C6alkenyl and C2-C6-alkynyl moieties in the aforementioned radicals R1 and R2 together with said carbon atom may form a carbonyl group, and wherein the C3-C8-cycloalkyl, cycloalkenyl, phenyl, naphthyl and heterocyclyl moieties in the aforementioned radicals R1 and R2 are unsubstituted or carry at least one substituent selected from hydroxy, halogen, cyano, nitro, Ci-C6-alkyl, C3-Ce-cycloalkyl, CrCehaloalkyl, CpCe-alkoxy, CrCe-haloalkoxy, CrCe-alkylamino, di-CrC6-alkylamino, Cr C6-alkylsulfonyl, phenyl and 5- or 6- membered hetaryl and/or wherein two radicals bound to the same carbon atom of said C3-Ce-cycloalkyl, C3-C8cycloalkenyl and heterocyclyl moieties of the radicals R1 and R2 together with said carbon atom may form a carbonyl group, and wherein
Rf and R9 are independently from each other selected from H, CrC6-alkyl, Ci-Cehaloalkyl, C3-C8-cycloalkyl, C3-C8-cycloalkenyl and heterocyclyl or
Rr and R9 together with the nitrogen atom to which they are bound form a cyclic amine, which may comprise a further heteroatom selected from O, N and S as a ring member;
n is an integer selected from 0,1, 2 or 3; and
R3 if présent are selected independently from each other from halogen, CrCe-alkyl, Cr C6-haloalkyl, CrC6-alkoxy, Ci-C6-haloalkoxy and C3-C0-cycloalkyl.
Surprisingly it has been found that the compounds of formula (la) or (Ib) according to the présent invention hâve significant CRTH2 antagoniste activity. Further it has been found that said compounds generally hâve enhanced chemical stability, enhanced pharmacokînetic properties (PK) and/or enhanced activity in a whole cell assay.
-716479
8/64
Thus the pyrazole compounds of formula (la) or (Ib) according to the présent invention are suitable for the prévention and/or treatment of diseases related to CRTH2-activity.
Accordingly the présent invention further relates to the use of pyrazole compounds of formula (la) or (Ib) according to the présent invention as médicaments.
Furthermore the présent invention relates to the use of compounds of formula (la) or (Ib) for preparing a médicament for the treatment of diseases related to CRTH2-activity. More specifically the présent invention relates to the use of pyrazole compounds of formula (la) or (lb) for preparing a médicament for the prévention and/or treatment of inflammatory, infectious and immunoregulatory disorders, respiratory or gastrointestinal diseases or complaints, inflammatory diseases of the joints and allergie diseases of the nasopharynx, eyes, and skin.
The présent invention further relates to compounds of formula (la) or (Ib) according to the invention for treating and/or preveting diseases related to CRTH2-activity More specifically the présent invention relates to compounds of formula (la) or (Ib) for use as a médicament for treating diseases related to CRTH2-activity. More specifically the présent invention relates to pyrazole compounds of formula (la) or (Ib) for use as a médicament for the prévention and/or treatment of inflammatory, infectious and immunoregulatory disorders, respiratory or gastrointestinal diseases or complaints, inflammatory diseases of the joints and allergie diseases of the nasopharynx, eyes, and skin.
Furthermore the présent invention relates to pharmaceutical formulations, containing one or more of the pyrazole compounds of formula (la) or (Ib) according to the présent invention as sole active substance or in combination with one or more active substances selected from among betamimetics, anticholinergics, corticosteroids, PDE4 inhibitors, LTD4 antagoniste, EGFR inhibitors, CCR3 antagoniste, CCR5 antagonists, CCR9 antagonists, 5-LO inhibitors, histamine-receptor antagonists, SYK inhibitors and sulphonamides.
The activity in a whole cell eosinophil shape change assay of the compounds of the invention can be determined, for example, according to the foliowing référencés: (i) Mathiesen JM, Ulven T, Martini L, Gerlach LO, Heinemann A, Kostenis E. Identification of indol dérivatives exclusively interfering with a G protein-independent signalling pathway of the prostaglandin D2 receptor CRTH2. Mol Pharmacol. 2005 Aug;68(2):393-402; (ii) Schulîgoi R, Schmidt R,
-816479
9/64
Geisslinger G, Kollroser M, Peskar BA, Heinemann A. PGD2 metabolism in plasma: kinetics and relationship with bioactivity on DP1 and CRTH2 receptors. Biochem Pharmacol. 2007 Jun 30;74(1 ):107-17; (iii) Royer JF, Schratl P, Carrillo JJ, Jupp R, Barker J, Weyman-Jones C, Beri R, Sargent C, Schmidt JA, Lang-Loidolt D, Heinemann A. A novel antagonïst of prostaglandin D2 blocks the locomotion of eosinophils and basophils. Eur J Clin Invest. 2008 Sep;38(9):663-71.
The chemical stability of the compounds of the invention can be determined, for example, under the following conditions: (i) 3 days incubation at 60°C in 0.1 N HCl (hydrolytic stability under acide conditions); (ii) 3 days incubation at 60°C in pH 4.0 buffer solution (hydrolytic stability under weakly acidic conditions); (iii) 3 days incubation at 60°C in pH 7.4 buffer solution (hydrolytic stability at physiological pH); (iv) 3 days incubation at 20°C in 0.3 % hydrogen peroxide (stability against oxidants); (v) 24 h incubation under UV-radiation (lambda = 300 - 800 nm, P = 250 W/m2) in water (stability against light). The kinetics of dégradation can, for example, be determined by HPLC analysis.
The pharmacokïnetic properties (PK) of the compounds of the invention can be determined in pre-clinical animal species, for example, mouse, rat, dog, guinea pig, mini pig, cynomolgus monkey, rhésus monkey. The pharmacokïnetic properties of a compound can be described, for example, by the following parameters: Mean résidence time, half-life, volume-ofdistribution, AUC (area under the curve), clearance, bioavailability after oral administration.
USED TERMS AND DEFINITIONS
Terms not specifically defined herein should be given the meanings that would be given to them by one of skill in the art in light of the disclosure and the context. As used in the spécification, however, unless specified to the contrary, the following terms hâve the meaning indicated and the following conventions are adhered to.
In the groups, radicals or moieties defined below, the number of carbon atoms is often specified preceding the group. As an example Ci-Ce-alkyl means an alkyl group or radical having 1 to 6 carbon atoms.
In general, for groups comprising two or more subgroups, the last named group is the radical attachment point.
-916479
10/64
Unless otherwise specified, conventional définitions of terms control and conventional stable atom valences are presumed and achieved in ail formulas and groups.
In general ail tautomeric forms and isomeric forms and mixtures, whether individual géométrie isomers or optical isomers or racemic or non-racemic mixtures of isomers of a chemical structure or compound, are comprised, unless the spécifie stereochemistry or isomeric form is speeifically indicated in the compound name or structure.
The term substituted as used herein, means that any one or more hydrogens on the designated atom, moiety or radical is replaced with a sélection from the indicated group of radicals, provided that the designated atom's normal valence is not exceeded, and that the substitution results in a stable compound.
The compounds disclosed herein can exist as pharmaceutically acceptable salts. The présent invention includes compounds in the form of salts, including acid addition salts. Suitable salts include those formed with both organic and inorganic acids. Such acid addition salts will normally be pharmaceutically acceptable. However, salts of non-pharmaceutically acceptable salts may be of utility in the préparation and purification of the compound in question. Basic addition salts may also be formed and be pharmaceutically acceptable. For a more complété discussion of the préparation and sélection of salts, refer to Pharmaceutical Salts: Properties, Sélection, and Use (Stahl, P. Heinrich. Wiley-VCH, Zurich, Switzerland, 2002).
The term pharmaceutically acceptable sait, as used herein, represents salts or zwitterionic forms ofthe compounds disclosed herein which are water oroil-soluble or dispersible and pharmaceutically acceptable as defined herein. The salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound in the form of the free base with a suitable acid. Représentative acid addition salts include acetate, adipate, alginate, L-ascorbate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, butyrate, camphorate, camphor sulfonate, citrate, digluconate, formate, fumarate, gentîsate, glutarate, glycérophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethansulfonate (isethionate), lactate, maleate, malonate, DL-mandelate, mesitylene sulfonate, methane sulfonate, naphthylene sulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, υνζ'
-1016479
11/64 pectinate, persulfate, 3-phenylproprionate, phosphonate, picrate, pivalate, propionate, pyroglutamate, succinate, sulfonate, tartrate, L-tartrate, trichloroacetate, trifluoroacetate, phosphate, glutamate, bicarbonate, para-toluenesulfonate (p-tosylate), and undecanoate. Also, basic groups in the compounds disclosed herein can be quaternized with methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides; dimethyl, diethyl, dibutyl, and diamyl sulfates; decyl, lauryl, myristyl, and steryl chlorides, bromides, and iodides; and benzyl and phenethyl bromides. Examples of acids which can be employed to form therapeutically acceptable addition salts include inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid and phosphoric acid, and organic acids such as oxalic acid, maleic acid, succinic acid and citrïc acid. Salts can also be formed by coordination of the compounds with an alkali métal or alkaline earth ion. Hence, the présent invention comprises sodium, potassium, magnésium, and calcium salts of the compounds disclosed herein, and the like.
Basic addition salts can be prepared during the final isolation and purification of the compounds by reacting a carboxy group with a suitable base such as the hydroxide, carbonate, or bicarbonate of a métal cation or with ammonia or an organic primary, secondary, or tertiary amine. The cations of pharmaceutically acceptable salts include lithium, sodium, potassium, calcium, magnésium, and aluminum, as well as nontoxic quaternary amine cations such as ammonium, tétraméthylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, diethylamine, ethylamine, tributylamine, pyridine, Ν,Ν-dimethylaniline, N-methylpiperidine, N-methylmorpholine, dicyclohexylamine, procaine, dibenzylamine, Ν,Ν-dibenzylphenethylamine, 1-ephenamine, and Ν,Ν'-dibenzylethylenediamine. Other représentative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine and piperazine.
While it may be possible for the compounds of the présent invention to be administered as the raw chemical, it is also possible to présent them as a pharmaceutical formulation. Accordingly, provided herein are pharmaceutical formulations which comprise one or more of certain compounds disclosed herein, or one or more pharmaceutically acceptable salts, esters, prodrugs, amides, or solvatés thereof, together with one or more pharmaceutically acceptable carrier and optionally one or more other therapeutic ingrédients. The carrier(s) must be acceptable in the sense of being compatible with the other ingrédients of the formulation and not deleterious to the récipient thereof. Proper formulation is dépendent upon the route of administration chosen. Any ofthe well-known techniques, carriers and xfi/'
-1116479
12/64 excipients may be used as suitable and as understood in the art; e.g. in Remington's Pharmaceutical Sciences. The pharmaceutical compositions disclosed herein may be manufactured in any manner known în the art, e.g., by means of conventional mixing, dissolving, granulatrng, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
The term halogen as used herein dénotés a halogen substituent selected from fluoro, chloro, bromo or iodo.
The term CrCe-alkyl as used herein (including the alkyl moieties of CrCe-alkoxy, CrCe-alkylamino, di-CrCe-alkylamino, C-i-Ce-alkylthio and the like) dénotés branched and unbranched alkyl moieties with 1 to 6 carbon atoms attached to the remaining compound at any position of the alkyl chain. The term CrC4 -alkyl accordingly dénotés a branched or unbranched alkyl moiety with 1 to 4 carbon atoms. -alkyl is generally preferred. Examples of Ci-C6-alkyl include: methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, secbutyl, tert-butyl, n-pentyl, iso-pentyl, neo-pentyl or hexyl. Unless stated otherwise, the définitions propyl, butyl, pentyl and hexyl include ail the possible isomeric forms of the groups in question. Thus, for example, propyl includes n-propyl and iso-propyl, butyl includes isobutyl, sec-butyl and tert-butyl etc.
The term CrCe-haloalkyl as used herein (including the alkyl moieties of CrC6-haloalkoxy, CrCe-haloalkylamino, di-CrCe-haloalkylamino, CrCe-haloalkylthio and the like) dénotés branched and unbranched alkyl moieties with 1 to 6 carbon atoms wherein one or more hydrogen atoms are replaced by a halogen atom selected from among fluorine, chlorine or bromine, preferably fluorine and chlorine, particularly preferably fluorine. The term Ci-C4-haloalkyl accordingly dénotés branched and unbranched alkyl moieties with 1 to 4 carbon atoms, wherein one or more hydrogen atoms are replaced analogously to what was stated above. C^C^-haloalkyl is generally preferred. Preferred examples include: CH2F, CHF2 and CF3.
The term C2-C6-alkenyr as used herein (including the alkenyl moieties of other radicals) dénotés branched and unbranched alkenyl groups with 2 to 6 carbon atoms attached to the remaining compound at any position of the alkenyl chain and having at least one double bond. The term C2-C4-alkenyl accordingly dénotés branched and unbranched alkenyl moieties with 2 to 4 carbon atoms. Preferred are alkenyl moieties with 2 to 4 carbon atoms. fjy'
-1216479
13/64
Examples include: ethenyl or vinyl, propenyl, butenyl, pentenyl or hexenyl. Unless otherwise stated, the définitions propenyl, butenyl, pentenyl and hexenyl include ail possible isomeric forms of the moieties in question. Thus, for example, propenyl includes 1-propenyl and 2propenyl, butenyl includes 1-, 2-and 3-butenyl, 1-methyl-1 -propenyl, 1-methyl-2-propenyl etc.
The term C2-C6-alkynyr' as used herein (including the alkynyl moieties of other radicals) dénotés branched and unbranched alkynyl groups with 2 to 6 carbon atoms attached to the remaining compound at any position of the alkynyl chain and having at least one triple bond. The term C2-C4-alkynyl accordingly dénotés branched and unbranched alkynyl moieties with 2 to 4 carbon atoms. Alkynyl moieties with 2 to 4 carbon atoms are preferred. Examples include: ethynyl, propynyl, butynyl, pentynyl, or hexynyl. Unless stated otherwise, the définitions propynyl, butynyl, pentynyl and hexynyl include ail the possible isomeric forms of the respective moieties. Thus, for example, propynyl includes 1-propynyl and 2-propynyl, butynyl includes 1-, 2- and 3-butynyl, 1-methyl-1-propynyl, 1-methyl-2-propynyl etc.
The term C3-C8-cycloalkyl as used herein (including the cycloalkyl moieties of other radicals) dénotés cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycîoheptyl and cyclooctyl. Preferred are cyclic alkyl groups with 3 to 6 carbon atoms, such as cyclopropyl, cyclopentyl and cyclohexyl.
The term C3-C8“Cycloalkenyl as used herein (including the cycloalkenyl moieties of other radicals) dénotés carbocyclic radicals having 3 to 8 carbon atoms and containing at least one, preferabiy one or two, non-conjugated double bonds. Examples are cyclopentenyl, cyclopantadienyl, cyclohexenyl and cyclohexadienyl.
The term heterocyclyl as used herein (including the heterocyclyl moieties of other radicals) dénotés 5- to 7-membered heterocyclic radicals and 5-to10-membered, bicyclic heterocyclic radicals, containing one, two or three heteroatoms, selected from O, N and S as ring members. The heterocyclyl may be linked to the molécule by a carbon atom or, if présent, by a nitrogen atom. The term heterocyclyl as used herein encompasses saturated or partially unsaturated heterocyclyl as well as hetaryl.
The term saturated or partially unsaturated heterocyclyl as used herein (including the heterocyclyl moieties of other radicals) dénotés 5- to 7-membered monocyclic heterocyclic
-1316479
14/64 radicals as defined above containing a number of double bonds such that no aromatic system is formed as well as 5- to 10-membered bicyclic heterocyclic radicals as defined above containing a number of double bonds such that no aromatic system is formed in at least one of the cycles.
Examples of monocyclic saturated or partially unsaturated heterocyclyl include pyrrolîdine, tetrahydrofurane, tetrahydrothiophene, thiazolidine, dioxolane, piperidine, tetrahydropyrane, tetrahydrothiopyrane, piperazine, morpholine, thiomorpholine, oxazepane, and the like.
Examples of bicyclic saturated or partially unsaturated heterocyclyl include dihydropyrrolizine, pyrrolizine, tetrahydroquinoline, tetrahydroisoquinoline, tetrahydroimidazopyridine, tetrahydropyrazolopyridine, benzopyrane, benzodiazépine, and the like.
The term hetaryl as used herein (including the heterocyclyl moieties of other radicals) dénotés 5- to 7-membered monocyclic heterocyclic radicals as defined above containing a number of double bonds such that an aromatic system is formed as well as 5- to 10membered bicyclic heterocyclic radicals as defined above containing a number of double bonds such that an aromatic system is formed in both cycles.
Examples of monocyclic aromatic heterocyclyl include furan, thiazole, pyrrole, thiophene, pyrazole, imidazole, thiadiazole, 1,2,3-triazole, 1,2,4-triazole, tetrazole, oxazole, oxadiazole, pyridine, pyridazine, pyrimidine, pyrazine, and the like.
Examples of bicyclic aromatic heterocyclyl include pyrrolizine, indol, indolizine, isoindol, indazol, purine, quinoline, isoquinoline, benzimidazol, benzofuran, benzothiazol, benzoisothiazol, pyridopyrimidine, pteridine, pyrimidopyrimidine, imidazopyridine, pyrazolopyridîne, and the like.
The term fused carbocyclic or heterocyclic moiety as used herein dénotés C3-Ce-cycloalkyl, C3-Ca-cycloalkenyl, benzene and heterocyclyl moieties as defined above, wherein said moieties share at least one bond with the cyclic moiety they are bound to. As an example benzene fused to benzene is naphthalene. Preferred are fused cyclic moieties sharing one bond with the cyclic moiety they are fused to. Further preferred the fused moiety is benzene.
-1416479
15/64
The term 3- to 8-membered ring formed by two radicals together with the carbon atom they are bound, wherein said ring may contain 1 or 2 heteroatoms selected from O, N and S as ring member as used herein dénotés C3-Ca-cycloalkyl, C3-C8-cycloalkenyl and heterocyclyl moieties as defined above.
The term cyclic amine formed by two radicals together with the nitrogen atom to which they are bound, wherein said ring may comprise a further heteroatom selected from O, N and S as a ring member as used herein dénotés cyclic amines having 3 to 8, preferably 5 or 6, ring members. Examples of such formed amines are pyrrolidine, piperidine, piperazîne, morpholine, pyrrol, imidazole, and the like.
The terms heterocyclyl-Ci-C6-alkyl, ^-Ca-cycloalkyl-CrCe-alkyl, phenyl-Ci-C6-alkyl and naphthyl-Ci-C6-alkyr as used herein dénoté alkyl moieties as defined above having 1 to 6 carbon atoms, wherein any one of the hydrogen atoms is replaced by a cyclic moiety as defined above. In these terms the alkyl moiety preferably has 1 to 4 carbon atoms (C1-C4alkyl). More preferably the alkyl moiety is methyl or ethyl, and most preferred methyl. Preferred examples of phenyl-CrC6-alkyl are benzyl or phenethyl.
The terms heterocyclyl-C2-C6-alkenyr, C3-Ce-cycloalkyl-C2-C6-alkenyl, phenyl-C2-C6alkenyl and naphthyl-C2-C6-alkenyl as used herein dénoté alkenyl moieties as defined above having 2 to 6 carbon atoms, wherein any one of the hydrogen atoms is replaced by a cyclic moiety as defined above. In these terms the alkenyl moiety preferably has 2 to 4 carbon atoms (C2-C4-alkenyl). More preferably the alkenyl moiety is ethenyl. A preferred example of phenyl-C2-C6-alkenyl is phenethenyl.
The spécifie and preferred définitions given for the indivîdual radicals and moieties Ra, Rb, Rc, Rd, Y1, Y2, Y3, Y4, Y5, Z, R1, R2, n and R3 herein below are valuable on their own as well as in combination. As will be understood preferred are compounds of formula (la) or (Ib) wherein one ore more ofthe indivîdual radicals and moieties Ra, Rb, Rc, Rd, Y1, Y2, Y3, Y4, Y5, Z, R1, R2, n and R3 hâve one of the meanings indicated as preferred herein-below and wherein the remaining radicals and moitiés are as specified hereinbefore. Most preferred are compounds of formula (la) or (Ib) wherein ail of the indivîdual radicals and moieties Ra, Rb, Rc, Rd, Y1, Y2, Y3, Y4, Y5, Z, R’, R2, n and R3 hâve one ofthe meanings indicated as preferred herein-below.
-1516479
16/64
One particular embodiment of the invention relates to pyrazole compounds of formula (la), wherein the individual moieties hâve one of the meanings given in the spécification. Preferred are compounds of formula (la), wherein the individual moieties hâve one of the preferred meanings given in the spécification.
Another particular embodiment of the invention relates to pyrazole compounds of formula (lb), wherein the individual moieties hâve one of the meanings given in the spécification. Preferred are compounds of formula (lb), wherein the individual moieties hâve one ofthe preferred meanings given in the spécification.
Preferred are pyrazole compounds of formula (la) or (lb), wherein Ra and Rb are independently selected hydrogen, CrCe-alkyl, Ci-C6 haloalkyl and C3-C8-cycloalkyl.
Particularly preferred are pyrazole compounds of formula (la) or (lb), wherein Ra and Rb are both hydrogen.
Likewise preferred are pyrazole compounds of formula (la) or (lb), wherein Rc and Rd are independently selected hydrogen, CrC6-alkyl, CrCo-haloalkyl and C3-Cfl-cycloalkyl.
Particularly preferred are pyrazole compounds of formula (la) or (lb), wherein Rc and Rd are both hydrogen.
Likewise preferred are pyrazole compounds of formula (la) or (lb), wherein Y1 is CRy1 or N, wherein R*1 has one of the meanings given for R*.
More preferred are pyrazole compounds of formula (la) or (lb), wherein Y1 is CRy1, in particular wherein Ry1 is selected from H, Ci-C6-alkyl, CrC6-alkoxy-CrC6-alkyl and Ci-C6haloalkyl.
Likewise preferred are pyrazole compounds of formula (la) or (lb), wherein Y2 is CR1'2, Y3 is CR*3, Y4 is CRy4 and/or Y5 is CR*5, wherein R*2, Ry3, Ry4 and R*5 independently from each other hâve one of the meanings as defined for Ry.
More preferred are pyrazole compounds of formula (la) or (lb), wherein Y2 is CR*2, Y3 is CR*3, Y4 is CR*4 and Y5 is CR*5, wherein R*2, R*3, R*4 and R*5 independently from each other
-1616479
17/64 hâve one of the meanings as defined for R¥, in particular wherein R1*2, Ry3, Ry4 and Ry5 are independently selected from H, halogen, CrCg-alkoxy, Ci-C6-alkoxy-CrC6-alkoxy and CrC6haloalkoxy.
One particular embodiment of the invention relates to pyrazole compounds of formula (la) or (Ib), wherein Z is O and the remaining moieties hâve one of the meanings given in the spécification, preferably one of the preferred meanings given in the spécification.
Another particular embodiment of the invetion relates to pyrazole compounds of formula (la) or (Ib), wherein Z is S and the remaining moieties hâve one of the meanings given in the spécification, preferably one of the preferred meanings given in the spécification.
Another particular embodiment of the invention relates to pyrazole compounds of formula (la) or (Ib), wherein Z is NRZ, wherein Rz is H, CrC6-alkyl or benzyl, and the remaining moieties hâve one of the meanings given in the spécification, preferably one of the preferred meanings given in the spécification.
Likewise preferred are pyrazole compounds of formula (la) or (Ib), wherein R1 and R2 independently from each other are selected from H, CrC6-alkyl, C3-Ca-cycloalkyl, phenyl and naphthyl.
More preferred are pyrazole compounds of formula (la) or (Ib), wherein R1 and R2 independently from each other are selected from H, CrC4-alkyl, C3-C6-cycloalkyl and phenyl.
Particularly preferred are pyrazole compounds of formula (la) or (Ib), wherein R1 and R2 are selected from CrC4-alkyl.
Likewise preferred are pyrazole compounds of formula (la) or (Ib), wherein n is 0, 1, 2 or 3, in particular wherein n is 0 or 1.
Likewise preferred are pyrazole compounds of formula (la) or (Ib), wherein R3 if présent are independently selected from halogen, Ct-CG-alkoxy and CrC6-haloalkoxy.
More preferred are pyrazole compounds of formula (la) or (Ib), wherein R3 if présent are independently selected from halogen, in particular from F, Cl and Br. y\/--1716479
18/64
One preferred particular embodiment of the invention relates to pyrazole compounds selected from compounds of formula (la j,
wherein Z, R1, R2, R3, Ry1, R*2, Ry3, Ry4 and R*5 hâve one of the meanings given above and n is 0 or 1.
More preferred are pyrazole compounds (la j wherein at least one of the moieties Z, R1, R2, R3, Ry1, Ry2. R*3, Ry4 and R*5 hâve one of the preferred meanings given above.
Another preferred particular embodiment of the invention relates to pyrazole compounds selected from compounds of formula (Ib1),
wherein Z, R1, R2, R3, Ry1, Ry2, R*3, Ry4 and R1* hâve one of the meanings given above.
More preferred are pyrazole compounds (laj wherein at leastone of the moieties Z, R1, R2, R3, Ry1, R*2, Ry3, Ry4 and R^ hâve one of the preferred meanings given above.
A further embodiment of the présent invention relates to compounds of formula (la) or (Ib), wherein the compounds of formula (la) or (Ib) are présent in the form of the individual optical isomers, mixtures of the individual enantiomers or racemates, preferably in the form of the enantiomerically pure compounds.
-1816479
19/64
A further embodiment of the présent invention relates to compounds of formula (la) or (lb), wherein the compounds of formula (la) or (lb) are présent in the form of the acid addition salts thereof with pharmacologically acceptable acids as well as optionally in the form of the solvatés and/or hydrates.
PREPARATION
The compounds according to the invention may be obtained using methods of synthesis which are known to a person skilled in the art and described in the literature of organic synthesis. Preferably the compounds are obtained analogously to the methods of préparation explained more fully hereinafter, in particular as described in the experimental section.
Compounds of formula (la) according to the présent inventioncan be prepared according to scheme 1.
Scheme 1
Starting material I
Starting material II
-1916479
20/64
(Deprotection) (la)
According to scheme 1 the compounds of the présent invention can be prepared employing as starting material I (1H-pyrazol-4-yl)acetic acid dérivatives, which are substituted with substituents Ra, Rb, R1, R2 and with a carboxylic acid protecting group PG. These compounds can, in some cases, be obtained from commercial sources or can be prepared according to literature procedures, for example WO 2007/141267. Suitable protecting groups can be selected from T. W. Greene, Protective Groups in Organic Synthesis, Wiley, 3fd édition, 1999. Preferred protecting groups PG are methyl, ethyl, tert-butyl.
Intermediate I can be obtained by alkylation of starting material I with a suitable 4-nitrobenzyl compounds as starting material II, wherein LG is a suitable leaving group such as halogen, in particular Br, or mesylate, in the presence of a base. Suitable bases are inorganic bases such as carbonates, especially potassium carbonate. The reaction is preferably carried out in an organic solvent such as dimethylformamide, dimethylsulfoxid, acetonitrile, tetrahydrofuran, dichloromethane or a mixture of solvents. The reaction usually takes place within 1 to 48 hours. Preferred reaction températures are between 0°C and the boiling point of the reaction mixture. When R1 is different from R2, the alkylation reaction may yield a mixture of regioisomers. The individual isomers may be separated by methods which are known to a person skilled in the art, for example, chromatography over silica gel employing a suitable solvent or solvent mixtures, or préparative reversed phase chromatography, employing a suitable gradient of solvents, or trituration or crystallization from suitable solvents or solvent mixtures.
Amine intermediate II can be prepared from intermediate I by réduction of the nitro group, for instance by hydrogenolysis in the presence of a catalyst, such as palladium on carbon or Raney Nickel. The réaction is preferably carried out in an inert organic solvent, such as methanol, éthanol, acetic acid, ethyl acetate or a mixture of solvents. The reaction usually takes place within 1 to 48 hours. Preferred reaction températures are between 0°C and 50°C.
-2016479
21/64
Preferred reaction pressures are between atmospheric pressure and 100 bar. The réduction ofthe nitro group in intermediate II can also be carried out according to alternative methods described in J. March, Advanced Organic Chemistry, Wiley, 4111 édition, 1992, p. 1216-1217. Amide intermediate III can be prepared from amine intermediate II by coupling with a carboxylic acid (Starting Material III) in the presence of a coupling reagent, such as 2-(1Hbenzotriazole-l-yh-IJ.S.S-tetramethyluronium tetrafluoroborate (TBTU), and a base, such as diisopropylethylamine. The reaction is preferably carried out in an inert organic solvent, such as dimethylformamide, tetrahydrofuran, dichloromethane or a mixture of solvents. The reaction usually takes place within 1 to 48 hours. Preferred reaction températures are between 0°C and 30°C. The coupling of a carboxylic acid to the amino group of intermediate III can also be carried out according to alternative methods described in J. March, Advanced Organic Chemistry, Wiley, 4’h édition, 1992, p. 419-421. Alternatively, instead of the carboxylic acid (starting material III) and a coupling reagent, the corresponding acyl chloride or anhydride may be employed.
Compounds of formula (la) can be obtained from intermediate III by removal ofthe protecting group PG. In the case the hydroxycarbonyl group is protected by CH3 or C2H5, this conversion can be carried out under aqueous conditions in the presence of an inorganic base, such as NaOH or LiOH. The reaction is preferably carried out in water or a mixture of water with CH3OH, C2H5OH, tetrahydrofuran or dioxane. The reaction usually takes place within 1 to 48 hours. Preferred reaction températures are between 0°C and the boiling point of the reaction mixture. In the case that PG is tert-butyl, the deprotection can be carried out under acidic conditions, for instance with trifluoroacetic acid, hydrochloric acid or montmorillonit. When using trifluoroacetic acid, the réaction can be carried out in neat trifluoroacetic acid or in an inert solvent, such as dichloromethane. The réaction usually takes place within 1 to 48 hours. Preferred reaction températures are between 0qC and 30°C. The cleavage of the protecting group PG may also be carried out according to alternative methods described in J. March, Advanced Organic Chemistry, Wiley, 4lh édition, 1992, p. 378-383 or in T. W. Greene, Protective Groups in Organic Synthesis, Wiley, 3rd édition, 1999.
Compounds of formula (Ib) can be obtained following the procedure depicted in scheme I using starting material of formula III', wherein
-2116479
22/64
Starting material III'
Y1, Y2, Y3, Y4, Y5 and Z hâve one of the meanings indicated above, instead of starting material III.
INDICATIONS
The compounds of formula (la) or (lb) according to the présent invention are especially useful for manufacturing a médicament for the prévention and/or treatment of diseases wherein the activity of a CRTH2-receptor is involved.
One embodiment of the présent invention relates to the manufacturing of a médicament for the prévention and/or treatment of a wide variety of inflammatory, infectious, and immunoregulatory disorders, respiratory or gastrointestinal diseases or complaints, inflammatory diseases of the joints and allergie diseases of the nasopharynx, eyes, and skin. Such disorders diseases and complaints include asthma and allergie diseases, éosinophilie diseases, chronic obstructive pulmonary disease, infection by pathogenic microbes (which, by définition, includes viruses), as well as autoimmune pathologies, such as the rheumatoid arthritis and atherosclerosis.
Preferred is the manufacturing of a médicament for the prévention and/or treatment of inflammatory or allergie diseases and conditions, including allergie or ποη-allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal polyposis, chronic rhinosinusitis, acute rhinosinusitis, asthma, pédiatrie asthma, allergie bronchitis, alveolitïs, Farmer’s disease, hyperreactive airways, allergie conjunctivitis, bronchitis or pneumonitis caused by infection, e.g. by bacteria or viruses or helminthes or fungi or protozoons or other pathogens, bronchiectasis, adult respiratory distress syndrome, bronchial and pulmonary edema, bronchitis or pneumonitis or interstitial pneumonitis caused by different origins, e.g. aspiration, inhalation of toxic gases, vapors, bronchitis or pneumonitis or interstitial pneumonitis caused by heart faîlure, X-rays, radiation, chemotherapy, bronchitis or pneumonitis or interstitial pneumonitis associated with collagenosis, e.g. lupus erythematodes, systemic scleroderma, lung fibrosis, idîopathic pulmonary lung fibrosis (IPF),
-2216479
23/64 interstitial lung diseases or interstitial pneumonitis of different origin, including asbestosis, silicosis, m. Boeck or sarcoidosis, granulomatosis, cystic fïbrosis or mucoviscidosis, or a1antitrypsin deficiency, éosinophilie cellulites (e.g., Well's syndrome), éosinophilie pneumonias (e.g., Loeffler’s syndrome, chronic éosinophilie pneumonia), éosinophilie fasciitis (e. g., Shulman’s syndrome), delayed-type hypersensitivity, non-allergic asthma; exercise induced bronchoconstriction; chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bronchitis, cough, pulmonary emphysema; systemic anaphylaxis or hypersensitivity responses, drug allergies (e.g., to penicillin, cephalosporin), eosinophilia-myalgia syndrome due to the ingestion of contaminated tryptophane, insect sting allergies; autoimmune diseases, such as rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, systemic lupus erythematosus, myasthenia gravis, immune thrombocytopenia (adult ITP, néonatal thrombocytopenia, pédiatrie ITP), immune hemolytic anémia (auto-immune and drug induced), Evans syndrome (platelet and red cell immune cytopaenias), Rh disease of the newborn, Goodpasture’s syndrome (anti-GBM disease), Celiac, autoimmune cardiomyopathy juvénile onset diabètes; glomerulonephritis, autoimmune thyroiditis, Behcet's disease; graft rejection (e.g., in transplantation), including allograft rejection or graftversushost disease; inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis; spondyloarthropathies; scleroderma; psoriasis (including T-cell mediated psoriasis) and inflammatory dermatoses such as an dermatitis, eczema, atopie dermatitis, allergie contact dermatitis, urticaria; vasculitis (e. g., necrotizing, cutaneous, and hypersensitivity vasculitis); erythema nodosum; éosinophilie myositis, éosinophilie fasciitis, cancers with leukocyte infiltration of the skin or organs.
METHOD OF TREATMENT
Accordingly, the compounds of formula (la) or (lb) according to the présent invention are useful in the prévention and/or treatment of a wide variety of inflammatory, infectious, and immunoregulatory disorders and diseases. Such disorders and diseases include but are not limited to asthma and allergie diseases, chronic obstructive pulmonary disease, infection by pathogenic microbes (which, by définition, includes viruses), autoimmune pathologies such as the rheumatoid arthritis and atherosclerosis.
As an example, an instant compound of formula (la) or (lb) which inhibits one or more functions of a mammalian CRTH2 receptor (e. g., a human CRTH2 receptor) may be administered to inhibit (i.e., reduce or prevent) inflammation and bronchoconstriction. As a «a.
-2316479
24/64 resuit, one or more inflammatory processes, such as leukocyte émigration, adhesion, chemotaxis, exocytosis (e. g., of enzymes, growth factors, histamine, cytotoxic proteins), inflammatory mediator release, survival or prolifération of CRTH2 expressing cells is inhibited. For example, activation or recruitment of Th2 cells, mast cells, basophils and éosinophilie to inflammatory sites (e. g., in asthma or allergie rhinitis) can be inhibited according to the présent method.
In particular, the compounds of the following examples hâve activity in blocking the activation and migration of cells expressing the CRTH2 receptor using the appropriate CRTH2 agonists in the aforementioned assays.
Diseases or conditions of humans which can be treated with inhibitors of CRTH2 receptor function, include, but are not limited to inflammatory or allergie diseases and conditions, including allergie or non-allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal polyposis, chronic rhinosinusitis, acute rhinosinusitis, asthma, pédiatrie asthma, allergie bronchitis, alveolitis, Farmer’s disease, hyperreactive airways, allergie conjunctivitis, bronchitis or pneumonitis caused by infection, e.g. by bacteria or viruses or helminthes or fungi or protozoons or other pathogens, bronchiectasis, adult respiratory distress syndrome, bronchial and pulmonary edema, bronchitis or pneumonitis or interstitial pneumonitis caused by different origins, e.g. aspiration, inhalation of toxic gases, vapors, bronchitis or pneumonitis or interstitial pneumonitis caused by heart failure, X-rays, radiation, chemotherapy, bronchitis or pneumonitis or interstitial pneumonitis associated with collagenosis, e.g. lupus erythematodes, systemic scleroderma, lung fibrosis, idiopathic pulmonary lung fibrosis (IPF), interstitial lung diseases or interstitial pneumonitis of different origin, including asbestosis, silicosis, m. Boeck or sarcoidosis, granulomatosis, cystic fibrosis or mucoviscidosis, or a1-antitrypsin deficiency, éosinophilie cellulites (e.g. Well’s syndrome), éosinophilie pneumonias (e.g. Loeffler’s syndrome, chronic éosinophilie pneumonia), éosinophilie fasciitis (e.g. Shulman's syndrome), delayed-type hypersensîtivity, non-allergic asthma, exercise induced bronchoconstrîction; chronic obstructive pulmonary disease (COPD), acute bronchitis, chronic bronchitis, cough, pulmonary emphysema; systemic anaphylaxis or hypersensitivity responses, drug allergies (e. g., to penicillin, cephalosporin), eosinophilia-myaîgia syndrome due to the ingestion of contaminated tryptophane, insect sting allergies; autoimmune diseases, such as rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, systemic lupus erythematosus, myasthenia gravis, immune thrombocytopenia (adult ITP, néonatal thrombocytopenia, pédiatrie ITP), immune hemolytic anémia (auto
-2416479
25/64 immune and drug induced), Evans syndrome (platelet and red cell immune cytopaenias), Rh disease of the newborn, Goodpasture’s syndrome (anti-GBM disease), Celiac, autoimmune cardio-myopathy juvénile onset diabètes; glomerulonephritis, autoimmune thyroiditis, Behcet's disease; graft rejection (e.g. in transplantation), încluding allograft rejection or graftversus-host disease; inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis; spondyloarthropathies; scleroderma; psoriasis (încluding T-cell mediated psoriasis) and inflammatory dermatoses such as an dermatitis, eczema, atopie dermatitis, allergie contact dermatitis, urticaria; vasculîtis (e.g. necrotizing, cutaneous, and hypersensitivity vasculîtis); erythema nodosum; éosinophilie myositis, éosinophilie fasciitis; cancers with leukocyte infiltration of the skin or organs.
COMBINATIONS
The compounds of formula (la) or (Ib) according to the présent invention may be used on their own or in combination with other compounds of formula (la) or (Ib). The compounds of formula (la) or (Ib) may optionally also be combined with other pharmacologically active substances.
Such pharmacologically active substances useable in the pharmaceutical composition containing compounds of formula (la) or (Ib) of the présent invention may be selected from but are not limited to the classes consisting of ft2-adrenoceptor-agonists (short and longacting beta mimetics), anti-cholinergics (short and long-acting), anti-inflammatory steroids (oral and topical corticosteroids), dïssociated-glucocorticoidmimetics, PDE3 inhibitors, PDE4 inhibitors, PDE7 inhibitors, LTD4 antagonists, EGFR inhibitors, PAF antagonists, Lipoxin A4 dérivatives, FPRL1 modulators, LTB4-receptor (BLT1, BLT2) antagonists, histamine-receptor antagonists, PI3-kinase inhibitors, inhibitors of non-receptor tyrosine kinases as for example LYN, LCK, SYK, ZAP-70, FYN, BTK or ITK, inhibitors of MAP kinases as for example p38, ERK1, ERK2, JNK1, JNK2, JNK3 or SAP, inhibitors of the NF-κΒ signaling pathway as for example IKK2 kinase inhibitors, iNOS inhibitors, MRP4 inhibitors, leukotriene biosynthesis inhibitors as for example 5-Lipoxygenase (5-LO) inhibitors, cPLA2 inhibitors, Leukotriene A4 hydrolase inhibitors or FLAP inhibitors, non-steroidal anti-inflammatory agents (NSAIDs), DP1-receptor modulators, thromboxane receptor antagonists, CCR1 antagonists, CCR2 antagonists, CCR3 antagonists, CCR4 antagonists, CCR5 antagonists, CCR6 antagonists, CCR7 antagonists, CCR8 antagonists, CCR9 antagonists, CCR10 antagonists, CXCR1 antagoniste, CXCR2 antagonists, CXCR3 antagonists, CXCR4 antagonists, CXCR5
-2516479
26/64 antagonists, CXCR6 antagonists, CX3CR1 antagonists, neurokinin (NK1, NK2) antagoniste, sphingosine 1-phosphate receptor modulators, sphingosine 1-phosphate-lyase inhibitors, Adenosine receptor modulators as for example A2a-agonists, modulators of purinergic receptors as for example P2X7 inhibitors, Histone Deacetylase (HDAC) activators, Bradykinin (BK1, BK2) antagonists, TACE inhibitors, PPAR gamma modulators, Rho-kinase inhibitors, interleukin 1-beta converting enzyme (ICE) inhibitors, Toll-like receptor (TLR) modulators, HMG-CoA reductase inhibitors, VLA-4 antagonists, ICAM-1 inhibitors, SHIP agonists, GABAa receptor antagonist, ENaC-inhibitors, Melanocortin receptor (MC1 R, MC2R, MC3R, MC4R, MC5R) modulators, CGRP antagonists, Endothelin antagonists, mucoregulators, immunotherapeutic agents, compounds against swelling of the airways, compounds against cough, CB2 agonists, retinoids, immunosuppressants, mast cell stabilizers, methylxanthine, opioid receptor agonists, laxatives, anti-foaming agents, antispasmodic agents, 5-HT4 agonists but also combinations of two or three active substances.
Preferred are combinations of two or three active substances, i.e.: CRTH2 antagonists according to the présent invention with betamimetics, anticholinergics, corticosteroids, PDE4 inhibitors, LTD4 antagonists, EGFR inhibitors, CCR3 antagonists, CCR5 antagonists, CCR9 antagonists, 5-LO inhibitors, histamine receptor antagonists, SYK inhibitors and sulfonamides, or i.e.:
• CRTH2 antagonists with betamimetics and corticosteroids, PDE4 inhibitors, CCR3 antagonists or LTD4 antagonists, • CRTH2 antagonists with anticholinergics and betamimetics, corticosteroids, PDE4 inhibitors, CCR3 antagonists or LTD4 antagonists, • CRTH2 antagonists with corticosteroids and PDE4 inhibitors, CCR3 antagonists or LTD4 antagonists • CRTH2 antagonists with PDE4 inhibitors and CCR3 antagonists or LTD4 antagonists
In the pharmaceutical compositions according to the présent invention the CRTH2 antagonists of formula (la) or (Ib) may be contained in a form selected from tautomers, optical isomers, enantiomers, racemates, dîastereomers, pharmacologically acceptable acid addition salts, solvatés or hydrates, as far as such forms exist, depending on the individual compound. Pharmaceutical compositions comprising one or more, preferably one, compound 1 in form of a substantially pure enantiomer are preferred.
-26i
27/64
In the pharmaceutical compositions according to the présent invention more than one CRTH2 antagonist of formula (la) or (Ib) and more than one further pharmacologîcally active compound can be présent.
PHARMACEUTICAL FORMS
Suitable préparations for administering the compounds of formula (la) or (Ib) include for example tablets, capsules, suppositories, solutions and powders etc. The content of the pharmaceutically active compound(s) should be in the range from 0.05 to 90 wt.-%, preferably 0.1 to 50 wt.-% of the composition as a whole.
Suitable tablets may be obtained, for example, by mixing the active substance(s) with known excipients, for example inert diluents such as calcium carbonate, calcium phosphate or lactose, disintegrants such as corn starch or alginic acid, binders such as starch or gélatine, lubricants such as magnésium stéarate or talc and/or agents for delaying release, such as carboxymethyl cellulose, cellulose acetate phthalate, or polyvinyl acetate. The tablets may also comprise several layers.
Coated tablets may be prepared accordingly by coating cores produced analogously to the tablets with substances normally used for tablet coatings, for example collidone or shellac, gum arable, talc, titanium dioxide or sugar. To achieve delayed release or prevent incompatibilities the core may also consist of a number of layers. Similarly the tablet coating may consist of a number or layers to achieve delayed release, possibly using the excipients mentioned above for the tablets.
Syrups or élixirs containing the active substances or combinations thereof according to the invention may additionally contain a sweetener such as saccharine, cyclamate, glycerol or sugar and a flavor enhancer, e.g. a flavoring such as vanillin or orange extract. They may also contain suspension adjuvants or thickeners such as sodium carboxymethyl cellulose, wetting agents such as, for example, condensation products of fatty alcohols with ethylene oxide, or preservatives such as p-hydroxybenzoates.
Solutions are prepared in the usual way, e.g. with the addition of isotonie agents, preservatives such as p-hydroxybenzoates or stabilizers such as alkali métal salts of ethylenediaminetetraacetic acid, optionally using emulsifiers and/or dispersants, while if%</
-27t
28/64 water is used as diluent, for example, organic solvents may optionally be used as solubilisers or dissolving aids, and the solutions may be transferred into injection vials or ampoules or infusion bottles.
Capsules containing one or more active substances or combinations of active substances may for example be prepared by mixing the active substances with inert carriers such as lactose or sorbitol and packing them into gélatine capsules.
Suitable suppositories may be made for example by mixing with carriers provided for this io purpose, such as neutral fats or polyethyleneglycol or the dérivatives thereof.
Excipients which may be used include but are not limited to water, pharmaceutically acceptable organic solvents such as paraffins (e.g. petroleum fractions), vegetable oils (e.g. groundnut or sesame oil), mono- or polyfunctional alcohols (e.g. éthanol or glycerol), carriers 15 such as e.g. natural minera! powders (e.g. kaolins, clays, talc, chalk), synthetic minerai powders (e.g. highly dispersed silicic acid and silicates), sugars (e.g. cane sugar, lactose and glucose), emulsifiers (e.g. lignin, spent sulphite liquors, methylcellulose, starch and polyvinylpyrrolidone) and lubricants (e.g. magnésium stéarate, talc, stearic acid and sodium lauryl sulphate).
For oral use the tablets may obviously contain, in addition to the carriers specified, additives such as sodium citrate, calcium carbonate and dicalcium phosphate together with various additional substances such as starch, preferably potato starch, gélatine and the like.
Lubricants such as magnésium stéarate, sodium laurylsulphate and talc may also be used to 25 produce the tablets. In the case of aqueous suspensions the active substances may be combined with various flavor enhancers or colorings in addition to the abovementioned excipients.
The compounds of formula (la) or (lb) may also be administered as préparations or
3o pharmaceutical formulations suitable for inhalation. Inhalable préparations include inhalable powders, propellant-containing metered-dose aérosols or propellant-free inhalable solutions. Within the scope of the présent invention, the term propellant-free inhalable solutions also include concentrâtes or stérile inhalable solutions ready for use. The formulations which may be used within the scope of the présent invention are described in more detail in the next part 35 of the spécification.
-2816479
29/64
The inhalable powders which may be used according to the invention may contain (la) or (Ib) either on its own or in admixture with suitable physiologically acceptable excipients.
If the active substances (la) or (Ib) are présent in admixture with physiologically acceptable excipients, the following physiologically acceptable excipients may be used to préparé these inhalable powders according to the invention: monosaccharides (e.g. glucose or arabinose), disaccharides (e.g. lactose, saccharose, maltose), oligo- and polysaccharides (e.g. dextrans), polyalcohols (e.g. sorbitol, mannitol, xylitol), salts (e.g. sodium chloride, calcium io carbonate) or mixtures of these excipients. Preferably, mono- or disaccharides are used, while the use of lactose or glucose is preferred, particularly, but not exclusively, in the form of their hydrates. For the purposes of the invention, lactose is the particularly preferred excipient, while lactose monohydrate is most particularly preferred.
Within the scope of the inhalable powders according to the présent invention the excipients hâve a maximum average particle size of up to 250 pm, preferably between 10 and 150 pm, most preferably between 15 and 80 pm. It may sometimes seem appropriate to add finer excipient fractions with an average particle size of 1 to 9 pm to the excipient mentioned above. These finer excipients are also selected from the group of possible excipients listed
2o hereinbefore. Finally, in order to préparé the inhalable powders according to the invention, micronised active substance 1, preferably with an average particle size of 0.5 to 10 pm, more preferably from 1 to 5 pm, is added to the excipient mixture. Processes for producing the inhalable powders according to the invention by grinding and micronising and finally mixing the ingrédients together are known from the prior art.
The inhalable powders according to the invention may be administered using inhalers known from the prior art.
The inhalation aérosols containing propellant gas according to the invention may contain the 30 compounds of formula (la) or (Ib) dissolved in the propellant gas or in dispersed form. The compounds of formula (la) or (Ib) may be contained in separate formulations or in a common formulation, in which the compounds of formula (la) or (Ib) are either both dissolved, both dispersed or in each case only one component is dissolved and the other is dispersed. The propellant gases which may be used to préparé the inhalation aérosols are known from the 35 prior art. Suitable propellant gases are selected from among hydrocarbons such as/l·'’—''
-2916479
30/64 n-propane, π-butane or isobutane and halohydrocarbons such as fluorinated dérivatives of methane, ethane, propane, butane, cyclopropane or cyclobutane. The abovementioned propellant gases may be used on their own or mixed together. Particularly preferred propellant gases are halogenated alkane dérivatives selected from TG134a and TG227 and 5 mixtures thereof.
The propellant-driven inhalation aérosols may also contain other ingrédients such as co-solvents, stabilizers, surfactants, antioxidants, lubricants and pH adjusters. Ail these ingrédients are known in the art.
The propellant-driven inhalation aérosols according to the invention mentioned above may be administered using inhalers known in the art (MDIs = metered dose inhalers).
Moreover, the active substances of formula (la) or (Ib) according to the invention may be administered in the form of propellant-free inhalable solutions and suspensions. The solvent used may be an aqueous or alcoholic, preferably an ethanolic solution. The solvent may be water on its own or a mixture of water and éthanol. The relative proportion of éthanol compared with water is not limited but the maximum is preferably up to 70 percent by volume, more particularly up to 60 percent by volume and most preferably up to 30 percent by volume. The remainder of the volume is made up of water. The solutions or suspensions containing compounds of formula (la) or (Ib) are adjusted to a pH of 2 to 7, preferably 2 to 5, using suitable acids. The pH may be adjusted using acids selected from înorganic or organic acids. Examples of particularly suitable înorganic acids include hydrochloric acid, hydrobromic acid, nitric acid, sulphuric acid and/or phosphoric acid. Examples of particularly suitable organic acids include ascorbic acid, citric acid, malic acid, tartaric acid, maleic acid, succinic acid, fumaric acid, acetic acid, formic acid and/or propionic acid etc. Preferred înorganic acids are hydrochloric and sulphuric acids. It is also possible to use the acids which hâve already formed an acid addition sait with one of the active substances. Of the organic acids, ascorbic acid, fumaric acid and citric acid are preferred. If desired, mixtures of the above acids may be used, particularly in the case of acids which hâve other properties in addition to their acidifying qualities, e.g. as flavorings, antioxidants or complexing agents, such as citric acid or ascorbic acid, for example. According to the invention, it is particularly preferred to use hydrochloric acid to adjust the pH. ----3016479
31/64
If desired, the addition of editic acid (EDTA) or one of the known salts thereof, sodium edetate, as stabilizer or complexing agent may be omitted in these formulations. Other embodiments may contain this compound or these compounds. In a preferred embodiment the content based on sodium edetate is less than 100 mg/100 ml, preferably less than
50mg/100ml, more preferably less than 20 mg/100 ml. Generally, inhalable solutions in which the content of sodium edetate is from 0 to 10 mg/100 ml are preferred.
Co-solvents and/or other excipients may be added to the propellant-free inhalable solutions. Preferred co-solvents are those which contain hydroxyl groups or other polar groups, e.g.
alcohols - particularly isopropyl alcohol, glycols - particularly propyleneglycol, polyethyleneglycol, polypropyleneglycol, glycolether, glycerol, polyoxyethylene alcohols and polyoxyethylene fatty acid esters. The terms excipients and additives in this context dénoté any pharmacologically acceptable substance which is not an active substance but which can be formulated with the active substance or substances in the physiologically suitable solvent in order to improve the qualitative properties of the active substance formulation. Preferably, these substances hâve no pharmacological effect or, in connection with the desired therapy, no appréciable or at least no undesirable pharmacological effect. The excipients and additives include, for example, surfactants such as soya lecithin, oleic acid, sorbitan esters, such as polysorbates, polyvinylpyrrolidone, other stabilizers, complexing agents, antioxidants and/or preservatives which guarantee or prolong the shelf life of the finished pharmaceutical formulation, flavorings, vitamins and/or other additives known in the art. The additives also include pharmacologically acceptable salts such as sodium chloride as isotonie agents.
The preferred excipients include antioxidants such as ascorbic acid, for example, provided that it has not already been used to adjust the pH, vitamin A, vitamin E, tocopherols and similar vitamins and provitamins occurring in the human body.
Preservatives may be used to protect the formulation from contamination with pathogens.
Suitable preservatives are those which are known in the art, particularly cetyl pyridinium chloride, benzalkonium chloride or benzoic acid or benzoates such as sodium benzoate in the concentration known from the prior art. The preservatives mentîoned above are preferably présent in concentrations of up to 50 mg/100 ml, more preferably between 5 and mg/100 ml.
-31·.
32/64
The dosage of the compounds according to the invention is naturally highfy dépendent on the method of administration and the complaint which is being treated. When adminîstered by inhalation the compounds of formula (la) or (lb) are characterized by a high potency even at doses in the pg range. The compounds of formula (la) or (lb) may also be used effectively above the pg range. The dosage may then be in the gram range, for example.
In another aspect the présent invention relates to the above-mentioned pharmaceutical formulations as such which are characterized in that they contain a compound of formula (la) or (lb), particularly the above-mentioned pharmaceutical formulations which can be adminîstered by inhalation.
The following examples of formulations illustrate the présent invention without restricting its scope:
Examples of pharmaceutical formulations:
A) Tablets per tablet active substance (la) or (lb) 100 mg lactose 140 mg maize starch 240 mg polyvinylpyrrolidone 15mg magnésium stéarate 5 mg
500 mg
The finely ground active substance, lactose and some of the maize starch are mixed together. The mixture is screened, then moistened with a solution of polyvinylpyrrolidone in water, kneaded, wet granulated and dried. The granules, the remaining maize starch and the magnésium stéarate are screened and mixed together. The mixture is pressed into tablets of suitable shape and size.
B) Tablets per tablet active substance (la) or (lb) 80 mg lactose 55 mg maize starch
-3216479
33/64
microcrystalline cellulose polyvinylpyrrolidone sodium carboxymethyl starch magnésium stéarate 35 mg 15 mg 23 mg 2 mg
Σ 400 mg
The finely ground active substance, some of the corn starch, lactose, microcrystalline cellulose and polyvinylpyrrolidone are mixed together, the mixture is screened and worked with the remaining corn starch and water to form a granulate which is dried and screened. The sodium carboxymethyl starch and the magnésium stéarate are added and mixed in and the mixture is compressed to form tablets of a suitable size.
C) Ampoule solution
active substance (la) or (lb) sodium chloride 50 mg 50 mg
water for inj. 5 ml
The active substance is dissolved in water at its own pH or optionally at pH 5.5 to 6.5 and sodium chloride is added to make the solution isotonie. The resulting solution is filtered to remove pyrogens and the filtrate is transferred under aseptie conditions into ampoules which are then sterilized and heat-sealed. The ampoules contain 5 mg, 25 mg and 50 mg of active substance.
D) Metering aérosol active substance (la) or (lb) sorbitan trioleate monofluorotrichloromethane and 0.005 0.1
TG134a:TG227 2:1 ad 100
The suspension is transferred into a conventional aérosol container with metering valve. Preferably 50 μΙ suspension are released on each actuation. The active substance may also be released in higher doses if desired (e.g. 0.02 wt.-%).
-33P01-2691-ff
34/64
E) Solutions (in mg/100ml) active substance (la) or (Ib) benzalkonium chloride 333.3 mg 10.0 mg
EDTA 50.0 mg
HCl (1N) ad pH 2.4
This solution can be prepared in the usual way.
F) Inhalable powder active substance (la) or (Ib) 12 pg lactose monohydrate ad 25 mg
The inhalable powder is prepared in the usual way by mixing the individual ingrédients.
The following examples serve to further illustrate the présent invention without restrîcting its scope.
EXAMPLES
I. HPLC METHODS
Method A:
HPLC-MS: AgilentHOO
Mobile phase:
A: water with 0.032% NH4OH
B: methanol
time in min %A %B flow rate in ml/min
0.00 95 5 1.50
2.00 0 100 1.50
2.50 0 100 1.50
2.60 95 5 1.50
2.90 95 5 1.50
Column: XBridge C18, 3,5pm, 4,6 x 50mm (column température: constant at 40°C).
Détection by diode array detector at 210-500 nm wavelength.
-3416479
35/64
Method B:
HPLC-MS: Waters ZQ MS, Alliance 2690/2695 HPLC, 2996 diode array detector
Mobile Phase:
A: water with 0.1% TFA
B: methanoi
time in min %A %B flow rate in ml/min
0.00 95 5 4.0
0.20 95 5 4.0
1.60 0 100 4.0
2.10 0 100 4.0
Column: Waters XBridge C18, 4.6 x 20 mm, 3.5 pm (column température: constant at 40°C). Détection by diode array detector at 210-400 nm wavelength.
Method C:
HPLC: Waters Acquity with DA and MS detector
Mobile Phase:
A: water with 0.1% TFA
B: methanoi
time in min %A %B flow rate in ml/min
0.00 99 1 1.5
0.05 99 1 1.5
1.05 0 100 1.5
1.20 0 100 1.5
Column: Waters XBridge BEH C18, 2.1 x 30 mm, 1.7 pm (column température: constant at 60°C). Détection by diode array detector at 210-400 nm wavelength.
Method D:
HPLC: Waters Acquity with DA and MS detector
Mobile Phase:
A: water with 0.13% TFA
B: methanoi with 0.05% TFA
time in min %A %B flow rate in ml/min
0.00 99 1 1.3
0.05 99 1 1.3
-3516479
36/64
1.05 0 100 1.3
1.20 0 100 1.3
Column: Waters XBridge BEH C18, 2.1 x 30 mm, 1.7 pm (column température: constant at 60°C). Détection by diode array detector at 210-400 nm wavelength.
Method E:
HPLC: Waters Acquity with DA and MS detector
Mobile Phase:
A: water with 0.1% TFA
B: methanol
time in min %A %B flow rate in ml/min
0.00 95 5 1.4
0.05 95 5 1.4
1.00 0 100 1.4
1.10 0 100 1.4
Column: Waters XBridge C18, 2.1 x 30 mm, 2.5 pm (column température: constant at 60°C). Détection by diode array detector at 210-400 nm wavelength.
Method F:
HPLC: Agilent 1200 with DA and MS detector
Mobile Phase:
A: water with 0.1 % TFA
B: methanol
time in min %A %B flow rate in ml/min
0.00 95 5 2.0
0.20 95 5 2.0
1.50 0 100 2.0
1.55 0 100 2.6
1.75 0 100 2.6
Column: Waters XBridge C18, 3 x 30 mm, 2.5 pm (column température: constant at 60°C).
Détection by diode array detector at 210-400 nm wavelength.
Method G:
HPLC-MS: Waters Alliance with DA and MS detector
Mobile Phase:
-3616479
37/64
A: water with 0.1% NH3
B: methanol with 0.1% NH3
time in min %A %B flow rate in ml/min
0.00 95 5 4.0
0.20 95 5 4.0
1.50 0 100 4.0
1.75 0 100 4.0
Column: Waters XBridge C18, 4.6 x 30 mm, 3.5 pm (column température: constant at 60°C). Détection by diode array detector at 210-400 nm wavelength.
Method H:
HPLC-MS: Waters Alliance with DA and MS detector
Mobile Phase:
A: water with 0.1% TFA
B: methanol
time in min %A %B flow rate in ml/min
0.00 95 5 4.8
1.60 0 100 4.8
1.85 0 100 4.8
1.90 95 5 4.8
Column: Waters SunFire C18, 4.6 x 30 mm, 3.5 pm (column température: constant at 60°C).
Détection by diode array detector at 210-400 nm wavelength.
Method J:
HPLC: Waters Acquity with DA and MS detector
Mobile Phase:
A: water with 0.13 % TFA
B: methanol with 0.05 % TFA
time in min %A %B flow rate in ml/min
0.00 99 1 1.2
0.15 99 1 1.2
1.10 0 100 1.2
1.25 0 100 1.2
Column: Waters Sunfire C18, 2.1 x 30 mm, 2.5 pm (column température: constant at 60°C).
Détection by diode array detector at 210-400 nm wavelength. U/—
-3716479
38/64
Method K:
HPLC-MS: Waters Alliance with DA and MS detector
Mobile Phase:
A: water with 0.1% TFA
B: methanol with 0.1% TFA
time in min %A %B flow rate in ml/min
0.00 95 5 4.0
0.20 95 5 4.0
1.50 0 100 4.0
1.75 0 100 4.0
1.85 95 5 4.0
Column: Waters XBridge C18, 4.6 x 30 mm, 3.5 pm (column température: constant at 60°C). Détection by diode array detector at 210-400 nm wavelength.
Method L:
HPLC-MS: Waters Alliance with DA and MS detector
Mobile Phase:
A: water with 0.1% TFA
B: methanol
time in min %A %B flow rate in ml/min
0.00 95 5 4.8
1.60 0 100 4.8
1.85 0 100 4.8
1.90 95 5 4.8
Column: Waters XBridge C18, 4.6 x 30 mm, 3.5 pm (column température: constant at 60°C). Détection by diode array detector at 210-400 nm wavelength.
Method M:
HPLC-MS: Waters 2695 HPLC, ZQ MS, 2996 diode array detector, 2695 autosampler
Mobile Phase:
A: water with 0.1% NH3
B: methanol with 0.1% NH3 time in min %A %B flow rate in ml/min
0.00 95 5 4.0
-3816479
39/64
0.20 95 5 4.0
1.50 0 100 4.0
1.75 0 100 4.0
Column: Waters XBridge C18, 4.6 x 30 mm, 3.5 pm (column température: constant at 60°C). Détection by diode array detector at 210-400 nm wavelength.
Method N:
HPLC: Waters Acquity with DA and MS detector
Mobile Phase:
A: water with 0.13% TFA
B: methanol with 0.08% TFA
time in min %A %B flow rate in ml/min
0.00 99 1 1.3
0.05 99 1 1.3
0.35 0 100 1.3
0.50 0 100 1.3
Column: Waters XBridge BEH C18, 2.1 x 30 mm, 1.7 pm (column température: constant at 60°C). Détection by diode array detector at 210-400 nm wavelength.
Method O:
HPLC: Agilent 1200 with DA and MS detector
Mobile Phase:
A: water with 0.1 % TFA
B: methanol
time in min %A %B flow rate in ml/min
0.00 95 5 1.9
0.20 95 5 1.9
1.55 0 100 1.9
1.60 0 100 2.4
1.80 0 100 2.4
Column: Waters XBridge C18, 3 x 30 mm, 2.5 pm (column température: constant at 60°C). Détection by diode array detector at 210-400 nm wavelength.
II. SYNTHESIS OF STARTING COMPOUNDS
-3916479
40/64
A) Synthesis of Amines
1. ) [1-(4-Aminobenzyl)-3,5-djmethyl-1H-pyrazol-4-yl]acetic acid methyi ester
a) To a solution of (3.5-dimethyl-1H-pyrazol-4-yl)-acetic acid methyl ester (3.90 g, 23 mmol) and 4-nitrobenzyl bromide (4.60 g, 20.7 mmol) in acetonitrile is added K2CO3 (2.76 g, 19.9 mmol) and the mixture is stirred for one hour at room température. The reaction mixture is poured into water and extracted twice with ethyl acetate. The organic phase is dried over MgSO4 and evaporated under reduced pressure. To yield 7.50 g of [3,5-Dimethyl-1 -(4-nitrobenzyl)-1H-pyrazol-4-yl]-aceticacid methyl ester (ESl mass spectrum: [M+H]* = 304).
b) To a solution of [3,5-dimethyl-1-(4-nitro-benzyl)-1H-pyrazol-4-yl]-acetic acid methyl ester (3.90 g, 10.3 mmol) in methanol (10 mL) is added 10 % palladium on charcoal (500 mg) and the mixture is hydrogenated. The catalyst is filtered off and the filtrate is concentrated under reduced pressure. The mixture is purified via préparative reversed phase HPLC (gradient of methanol in water + 0.1 % NH3) to yield 1.18 g of the title compound (ESl mass spectrum: [M+H]* = 274; Rétention time HPLC: 2.13 min (method A))
2. ) [1-(4-Amino-2-chloro-benzyl)-3,5-dimethyl-1H-pyrazol-4-yl]acetic acid ethyl ester
a) To a solution of (3,5-dimethyl-1H-pyrazol-4-yl)-acetic acid ethyl ester (1.40 g, 7.7 mmol) and 2-chloro-4-nitrobenzyl bromide (4.60 g, 20.7 mmol) in 20 mL acetonitrile is added K2CO3 (1.59 g, 11.5 mmol) and the mixture is stirred for 48 hours at room température. The solvent is removed by évaporation and the residue is dissolved in dichloromethane/water. After extraction with dichloromethane the organic layer is dried over Na2SO4 and evaporated under reduced pressure to yield 2.79 g of [3,5-dimethyl-1-(2-chloro-4-nitro-benzyl)-1H-pyrazol-4-yl]acetic acid ethyl ester (ESl mass spectrum: [M+H]* = 352; Rétention time: 1.95 min (method A).
b) To a solution [3,5-dimethyl-1-(2-chloro-4-nitro-benzyl)-1H-pyrazol-4-yl]acetic acid ethyl ester (2.39 g, 6.8 mmol) in methanol (40 mL) is added Raney nickel (250 mg) and the mixture is hydrogenated. The catalyst is filtered off and the filtrate is concentrated under ycs'
-4016479
41/64 reduced pressure to yield 1,18 g of the title compound (ESI mass spectrum: [M+Hf = 322;
Rétention time HPLC: 1.76 min (method A)).
3. ) [1-(4-Amino-benzyl)-3,5-diethyl-1H-pyrazol-4-yl]-acetic acid ethyl ester
a) To a solution of (3,5-diethyl-1H-pyrazol-4-yl)-acetic acid ethyl ester (10.95 g, 52.1 mmol) and 4-nitrobenzyl bromide (14.625 g, 68 mmol) in acetonitrile (110 mL) is added K2CO3 (10.80 g, 78.1 mmol) and the mixture is stirred for 48 hours at room température. The reaction mixture is poured into water and extracted twice with ethyl acetate. The organic phase is dried over MgSO4 and evaporated under reduced pressure. The residue is purified by MPLC with ethyl acetate/cyclohexane to yield 12.50 g [3,5-diethyl-1-(4-nitro-benzyl)-1Hpyrazol-4-yl]acetic acid ethyl ester (ESI mass spectrum: [M+H]+ = 346; Rétention time HPLC: 1.42 min (method B)).
b) To a solution of [3,5-diethyl-1-(4-nitro-benzyl)-1H-pyrazol-4-yl]acetic acid ethyl ester (6.66 g, 19.3 mmol) in methanol (500 mL) is added Raney nickel (500 mg) and the mixture is hydrogenated at 50 psi and room température. The catalyst is filtered off and the filtrate is concentrated under reduced pressure to yield 4.38 g of the title compound (ESI mass spectrum: [M+H]+ = 316; Rétention time HPLC: 1.09 min (method B)).
4. ) [1-(4“Amino-2-fluorobenzyl)“3,5-dimethyl-1H-pyrazol-4-yl]acetic acid methyl ester
a) To a solution of (3,5-dimethyl-1 H-pyrazol-4-yl)-acetic acid methyl ester (10 g, 48.9 mmol) and 2-fluoro-4-nitrobenzyl bromide (11.5 g, 49.1 mmol) in acetonitrile (150 mL) is added K2CO3 (10.1 g, 73.3 mmol) and the mixture is stirred for 48 hours at 60°C. The solvent is removed by évaporation and the residue is dissolved in dichloromethane/water. After extraction with dichloromethane the organic layer is dried over Na2SO4 and evaporated under reduced pressure. The residue is purified by MPLC (cyclohexane/diethyl ether 7:3) to yield 13 g of [3,5-dimethyl-1-(2-fluoro-4-nitro-benzyl)-1H-pyrazol-4-yl]acetic acid methyl ester (ESI mass spectrum: [M+Hf = 322; Rétention time (HPLC): 0.85 min (method C)).
b) To a solution of [3,5-dimethyl-1-(2-fluoro-4-nitro-benzyl)-1H-pyrazol-4-yl]acetic acid methyl ester (13 g, 40.4 mmol) in methanol (250 mL) is added Raney nickel (6 g) and the mixture is ,
-4116479
42/64 hydrogenated at 50 psi and 50°C. The catalyst is filtered off and the filtrate is concentrated under reduced pressure. The mixture is purified by crystailization in diisopropyl ether to yield 12.8 g of the title compound (ESl mass spectrum: [M+H]+ = 292; Rétention time HPLC: 0.61 min (method C)).
5. ) [1-(4-Amino-2-chloro-benzyl)-3,5-dimethyl-1H-pyrazol-4-yl]-acetic acid tert-butyl ester
a) To a solution of (3,5-dimethyl-1H-pyrazol-4-yl)-acetic acid tert-butyl ester (2.6 g, 12.4 mmol, préparation according to WO2007/141267 by using 2,4-pentanedione instead of 3,5heptanedione) and 2-chloro-4-nitrobenzyl bromide (3.11 g, 12.4 mmol) in acetonitrile (30 mL) is added K2CO3 (2.575 g, 18.6 mmol) and the mixture is stirred for 48 hours at room température and after that 2 hours at 60°C. The solid is filtered off and the solvent is removed by évaporation. The residue is dissolved in dichloromethane/water. After extraction with dichloromethane the organic layer is dried over MgSO4 and evaporated under reduced pressure to yield 4.4 g of [3,5-dimethyl-1-(2-chloro-4-nitrobenzyl)-1 H-pyrazol-4-yl]acetic acid tert-butyl ester (ESI mass spectrum: [M+H]+ = 380).
b) To a solution of [3,5-dimethyl-1-(2-chloro-4-nitrobenzyl)-1H-pyrazol-4-yl]acetic acid tertbutyl ester (4.40 g, 11.6 mmol) in methanol (80 mL) is added Raney nickel (440 mg) and the mixture is hydrogenated at 50 psi and room température for 12 hours. The catalyst is filtered off and the filtrate is concentrated under reduced pressure to yield 2.4 g of the title compound (ESl mass spectrum: [M+H]+ = 350; Rétention time HPLC: 0.76 min (method D)).
6. ) [1-(4-Aminobenzyl)-3,5-dimethyl-1H-pyrazol-4-yl]acetic acid tert-butyl ester
The title compound (ESl mass spectrum: [M+H]+ = 316; Rétention time HPLC: 0.65 min (method E)) is synthesized in analogy to procedure II.A.5 by using 4-nitrobenzylbromide instead of 2-fluoro-4-nitrobenzylbromide.
7. ) [1-(4-Amino-2-fluorobenzyl)-3,5-dimethyl-1H-pyrazol-4-yl]acetic acid tert-butyl ester
a) To a solution of (3,5-dimethyl-1 H-pyrazol-4-yl)acetic acid tert-butyl ester (10 g, 47.6 mmol, préparation according to WO2007/141267 by using 2,4-pentanedione instead of 3,5
-4216479
43/64 heptanedione) and 2-fluoro-4-nitrobenzyl bromide (11.2 g, 47.9 mmol) in acetonitrile (150 mL) is added K2CO3 (6.615 g, 47.9 mmol) and the mixture is stirred for 24 hours at room température. The solid is filtered off and the solvent is removed by évaporation. The residue is dissolved in dichloromethane/water. After extraction with dichloromethane the organic layer is dried over MgSO4 and evaporated under reduced pressure. The residue is purified by MPLC (cyclohexane/ethyl acetate 7:3, silicagel 60) to yield 13.6 g of [3,5-dimethyl-1-(2fluoro-4-nitrobenzyl)-1H-pyrazol-4-yl]acetic acid tert-butyl ester (ESl mass spectrum: [M+H]+ = 364 TLC: Rf = 0.23 (cyclohexan/ethyl acetate 7:3, solicagel 60 F254)).
b) To a solution of [3,5-dimethyl-1-(2-fluoro-4-nitrobenzyl)-1H-pyrazol-4-yl]acetic acid tertbutyl ester (13.6 g, 37.4 mmol) in methanol (250 mL) is added Raney nickel (6 g) and the mixture is hydrogenated at 50 psi and 50°C for 12 hours. The catalyst is filtered off and the filtrate is concentrated under reduced pressure to yield 11.6 g of the tiltle compound (ESl mass spectrum: [M+Hf = 334; TLC: Rf = 0.53 (dichloromethane/methanol 95:5, silicagel 60 F254)).
B) Synthesis of carboxylic acids
1.) 1-Ethyl-5-fluoro-1H-indole-2-carboxylic acid
a) To a solution of 5-fluoroindol-2-ethyl ester (200 mg, 0.965 mmol) in dimethylsulfoxide (6 mL) is added potassium tert-butylate (108 mg, 0.965 mmol) and the mixture is stirred at 50°C for 30 minutes. After cooling to room température bromoethane (0.081 mL, 1.06 mmol) is added and the mixture is stirred at room température for 2.5 hours. With cooling water is added and the mixture is extracted with ethyl acetate. The organic layer is washed with water and saturated aqueous NaCI solution, dried over MgSO4 and the solvent is evaporated in vacuo to yield 200 mg of 1-ethyl-5-fluoro-1H-indole-2-carboxylic acid ethyl ether (ESl mass spectrum: [M+Hf = 236).
b) To a solution of 1-ethyl-5-fluoro-1 H-indole-2-carboxylic acid ethyl ether (200 mg, 0.85 mmol) in dioxan (2 mL) is added an aqueous solution of NaOH (1 M, 1.7 mL) and the mixture is stirred at 60°C for 1 hour. After évaporation of the solvent the residue was suspended in a few water and neutralized with acetic acid (2 M). The precipitate is filtered, washed with
-4316479
44/64 water and dried to yield 140 mg of the title compound (ESI mass spectrum: [M+H]’ = 208;
Rétention time HPLC: 1.21 min (method F)).
The following indole carboxylic acids are prepared likewise in anlaogy to this method:
1-Benzyl-1H-indole-2-carboxylic acid (ESI mass spectrum: [M+H]* = 252 Rétention time HPLC: 0.84 min (method E));
1-Butyl-1H-indole-2-carboxylic acid (ESI mass spectrum: [M+H]* = 218);
5- Fluoro-1-propyl-1H-indole-2-carboxylic acid (ESI mass spectrum: [M+H]’= 222);
1-Butyl-5-fluoro-1H-indole-2-carboxylic acid (ESI mass spectrum: [M+H]+ = 236);
1-Propyl-1H-indole-2-carboxylic acid (ESI mass spectrum: [M+H]* = 204);
1-Ethyl-4-fluoro-1H-indole-2-carboxylîcacid (ESI mass spectrum: [M+H]* = 208; Rétention time HPLC: 0.80 min (method E);
1-Ethyl-6-fluoro-1H-indole-2-carboxylîc acid (ESI mass spectrum: [M+H]'=206; Rétention time HPLC: 0.71 min (method E));
6- Fluoro-1-propyl-1H-indole-2-carboxylic acid (ESI mass spectrum: [M-H] = 220; Rétention time HPLC: 0.77 min (method E)).
2.) 3-Ethyl-5-fluorobenzofuran-2-carboxylic acid
a) To a solution of 5-fluoro-2-hydroxy-propiophenone (0.9 g, 5.2 mmol) and tert-butyl bromoacetate (0.9 mL, 6.1 mmol) in acetonitrile (15 mL) is added K2CO3 (1.08 g, 7.8 mmol) and the mixture is refluxed for 3 hours. After cooling to room température the mixture is poured into water and extracted with ethyl acetate. The organic layer is washed twice with water, dried over MgSO4 and the solvent is evaporated in vacuo to yield 1.47 g of (4-fluoro-2propionylphenoxy)acetic acid tert-butyl ester (ESI mass spectrum: [M+H]* = 283; Rétention time HPLC: 0.88 min (method D)). v/*
-4416479
45/64
b) To a solution of (4-fluoro-2-propionylphenoxy)acetic acid tert-butyl ester (1.47 g, 5.2 mmol) in dry éthanol (20 mL) is added solution of sodium methanolat in methanol (5.4 M, 20 mL) and the mixture is stirred at 80°C for 12 hours. After cooling to room température and évaporation of the solvent the residue was dissolved in water and acidified with hydrochloric acid (1 M). The precipitate is filtered, washed with water and dried to yield 440 mg of the title compound (ESI mass spectrum: [M-H]' = 207; Rétention time HPLC: 0.87 min (method G)).
The following benzofuran carboxylic acids are prepared likewise in analogy to this method:
7-Chloro-3-methylbenzofuran-2-carboxylic acid (ESI mass spectrum: [M-H]' = 209; Rétention time HPLC: 1.28 min (method H));
5- Fluoro-3-propylbenzofuran-2-carboxylic acid (ESI mass spectrum: [M-H]’ = 221; Rétention time HPLC: 1.01 min (method G));
3-Ethyl-7-fluorobenzofuran-2-carboxylic acid (ESI mass spectrum: [M-H]' = 207; Rétention time HPLC: 0.77 min (method E));
3-Ethyl-5,7-difluorobenzofuran-2-carboxylic acid (ES! mass spectrum: [M-H]‘ = 225; Rétention time HPLC: 1.32 min (method L));
6- Chlorobenzofuran-2-carboxylic acid (ESI mass spectrum: [M-H]' = 195; Rétention time HPLC: 1.72 min (method H)).
III) SYNTHESIS OF COMPOUNDS (la) and (Ib)
Compound 1: (1-(2-fluoro-4-[(1H-indole-2-carbonyl)amino]benzyl)-3,5-dimethyl-1H-pyrazol-4yl)acetic acid (Coupling method C1):
a) To a solution of indole-2-carboxylic acid (80 mg, 0.50 mmol) in Ν,Ν-dimethylformamide (1.5 mL) are added TBTU (139 mg, 0.43 mmol) and Ν,Ν-diisopropyl amine (0.126 mL, 0.74 mmol). Subsequently [1-(4-amino-2-fluorobenzyl)-3,5-dimethyl-1H-pyrazol-4-yl]acetic acid methyl ester (120 mg, 0.41 mmol) is added. The mixture is stirred at room température for 12
-4516479
46/64 hours. After that an aqueous solution of K2CO3 (2 M, 0.5 mL) is added. The obtained mixture is flushed through AI2O3 with dichloromethane/methanol (9:1, 10 mL). The solvent is removed in vacuo to yield 82.3 mg of (1-(2-fluoro-4-[(1H-indole-2-carbonyl)amino]benzyl)-3,5-dimethyl1 H-pyrazol-4-yl)acetic acid methyl ester (ESl mass spectrum: [M+Hf = 435; Rétention time HPLC: 0.41 min (method N).
b) (1-(2-Fluoro-4-[(1H-indole-2-carbonyl)amino]benzyl)-3,5-dimethyl-1H-pyrazol-4-yl)acetic acid methyl ester (82 mg, 0.19 mmol) is dissolved in methanol (0.5 mL). An aqueous solution of NaOH (4M, 0.3 mL) is added and the mixture is stirred at room température for 2 hours. The resulting mixture is diluted with methanol/water, the solid is filtered off and after évaporation the residue is purified by HPLC (Gilson, XRS Pursuit, methanol/H2O+0.1 % conc. NH3). The fractions containing the title compound are concentrated and lyophilized to yield 15 mg of the title compound (ESl mass spectrum: [M+Hf = 421; Rétention time HPLC: 1.04 min (method G)).
1H-NMR 400 MHz (DMSO-d6): δ [ppm] = 2.03 (s, 3H). 2.14 (s, 3H), 2.52 (s, 3H), 3.21 (s, 2H), 5.16 (s, 2H), 6.99 (t, 1H), 7.08 (t, 1H), 7.23 (t, 1H), 7.39 (s. 1H), 7.48 (m, 2H), 7.66 (d, 1H), 7.79 (d, 1H), 10.45 (s, 1H), 11.79 (br., 1H).
Compounds 2 and 3 of table 1 below hâve likewise been prepared in analogy to coupling method C1 using suitable starting amines and carboxylic acids.
Compound 4: (1 -{4-[(5-Fluoro-3-methylbenzofuran-2-carbonyl)amino]benzyl)-3,5-dimethyl1 H-pyrazol-4-yl)acetic acid (Coupling method C2):
a) To a solution of [1-(4-aminobenzyl)-3,5-dimethyl-1 H-pyrazol-4-yl]acetic acid methyl ester (400 mg, 1.46 mmol) in 5 mL dichloromethane are added diisopropylethylamine (1.5 mL, 8.8 mmol) and 5-fluoro-3-methyl-1-benzofuran-2-carboxylic acid (369 mg, 1.9 mmol). After stirring at room température for 10 minutes a 50 % solution of 1-propylphosphonic acid cyclic anhydride in ethyl acetate (1.725 mL, 2.93 mmol) is added with cooling and the mixture is stirred at room température for 12 hours. The solvent is evaporated in vacuo and the residue is purified by MPLC (dichloromethane/methanol 98:2) to yield 410 mg of (1-{4-[(5-fluoro-3methylbenzofuran-2-carbonyl)amino]benzyl)-3,5-dimethyl-1 H-pyrazol-4-yl)acetic acid methyl ester (ESl mass spectrum: [M+Hf = 450; TLC: Rf = 0.56 (dichloromethane/methanol 95:5, silicagel 60 F254)).
-4616479
47/64
b) To a solution of (1-{4-[(5-fluoro-3-methylbenzofuran-2-carbonyl)amino]benzyl}-3,5dimethyl-1H-pyrazol-4-yl)acetic acid methyl ester (410 mg, 0.91 mmol) in dioxan/water (10 mL/10 mL) is added 1 M NaOH (2.3 mL) and the mixture is stirred at room température for 24 hours. The mixture is diluted with water and acidified with hydrochlorîc acid (1 M, 3.25 mL). The precipitate is filtered, washed with water and dried to yield 338 mg of the title compound (ESI mass spectrum: [M+H]+ = 436; Rétention time HPLC: 0.85 min (method D).
1H-NMR 400 MHz (DMSO-d6): δ [ppm] = 2.05 (s, 3H), 2.10 (s, 3H), 2.52 (s, 3H), 3.24 (s, 2H), 5.16 (s, 2H), 7.10 (d, 2H), 7.37 (t, 1H), 7.66 (m, 2H), 7.75 (d, 2H), 10.40 (s, 1H), 12.05 (br.,
1H).
Compounds 5 to 26 and 57 to 59 of tables 1 and 2 below hâve likewise been prepared in analogy to coupiing method C2 using suitable starting amines and carboxylic acids.
Compound 27: (1 -{2-Fluoro-4-[(5-fluoro-3-methylbenzofuran-2-carbonyl)amino]benzyl}-3,5dimethyl-1H-pyrazol-4-yl)-acetic acid (Coupiing method C3):
a) To a solution of 5-fluoro-3-methyl-1-benzofuran-2-carboxylicacid (194 mg, 1 mmol) in 5 mL dimethylformamide is added diisopropylethylamine (0.516 mL, 3 mmol) and HATU (399 mg, 1.05 mmol) is added. After stirring at room température for 25 minutes dimethylformamide (1 mL) and then intermediate [1-(4-amino-2-fluorobenzyl)-3,5-dimethyl1 H-pyrazol-4-yl]acetic acid methyl ester (291 mg, 1 mmol) is added. Subsequently diisopropylethylamine (0.344 mL, 2 mmol) and dimethylformamide (2 mL) is added, and the mixture is stirred at room température for 48 hours. Then ethyl actetate and water are added and the precipitate is filtered off. The organic layer is extracted twice with acetic acid (1 N), once with an aqueous solution of NaHCO3 (5 % by weight) and twice with water, dried over MgSO4. The solvent is removed by évaporation in vacuo. The residue is prurified by MPLC (dichloromethane/methanol 96:4) to yield 120 mg of (1-(2-fluoro-4-[(5-fluoro-3methylbenzofuran-2-carbonyl)-amino]-benzyl)-3I5-dimethyl-1H-pyrazol-4-yl)-acetic acid methyl ester (ESI mass spectrum: [M+H]+ = 468; TLC: Rf = 0.72 (dichloromethane/methanol 9:1, silicagel 60 F254)).
b) To a solution of (1-(2-fluoro-4-[(5-fluoro-3-methylbenzofuran-2-carbonyl)-amino]-benzyl)-
3,5-dimethyl-1 H-pyrazol-4-yl)-acetic acid methyl ester (119 mg, 0.26 mmol) in dioxan/water
-4716479
48/64 (7 mL/7 mL) is added an aqueous solution of NaOH (1 M, 2.3 mL) and the mixture is stirred at room température for 12 hours and at 60°C for 2 hours. The mixture is diluted with water, acidified with hydrochloric acid (1 M, 1 mL) and extracted with ethyl acetate. The organic layer is dried over MgSO41 the solvent evaporated in vacuo, the residue is crystallized with diisopropylether and the precipitate is isolated by filtration to yield 69 mg of the title compound (ESI mass spectrum: [M+Hf = 454; Rétention time HPLC: 0.90 min (method D)).
1H-NMR 400 MHz (DMSO-d6): δ [ppm] = 2.03 (s, 3H), 2.14 (s, 3H), 2.57 (s, 3H), 3.28 (s, 2H), 5.18 (s, 2H), 6.96 (t, 1H), 7.37 (t, 1H), 7.54 (d, 1H), 7.66 (m, 2H), 7.79 (d, 1H), 10.60 (s, 1H), 12.07 (br., 1H).
Compounds 28 to 31 of table 1 below hâve likewise been prepared in analogy to coupling method C3 using suitable starting amines and carboxylic acids.
Compounds 32: (1-(4-[(1-Ethyl-5-fluoro-1H-indole-2-carbonyl)amino]-2-fluorobenzyl)-3,5dimethyl-1H-pyrazol-4-yl)acetic acid (Coupling method C4):
a) To a solution of 1-ethyl-5-fluoro-1H-indole-2-carboxylic acid (140 mg, 0.68 mmol) in 6 mL dichloromethane is added oxalyl chloride (0.094 mL, 0.68 mmol) and a drop dimethylformamide. After stirring at room température for 1 hour the solvent is removed by évaporation in vacuo. The residue is dissolved in dichloromethane (5 mL) and dropped to a solution of [1-(4-amino-2-fluorobenzyl)-3,5-dimethyl-1H-pyrazol-4-yl]acetic acid methyl ester (177 mg, 0.61 mmol) and diisopropylethylamine (0.23 mL, 1.35 mmol) in dichloromethane (5 mL). After addition of 4-dimethylaminopyridine (8.255 mg, 0.069 mmol) the solution is stirred at room température for 12 hours. The solution is extracted twice with hydrchloric acid (1 M), twice with water, twice with an aqueous solution of NaOH (1 M) and twice with water. The organic layer is dried over MgSO4, filtered and the solvent is evaporated in vacuo. The residue is purified by MPLC (dichloromethane/methanol 95:5) to yield 160 mg of (1-(4-[(1ethyl-5-fluoro-1H-indole-2-carbonyl)amino]-2-fluorobenzyl)-3,5-dimethyl-1H-pyrazol-4yl)acetic acid methyl ester (ESI mass spectrum: [M+H]+ = 481; Rétention time HPLC: 0.93 min (method D)).
b) To a solution of (1-(4-[(1-ethyl-5-fluoro-1H-indole-2-carbonyl)amino]-2-fluorobenzyl)-3,5dimethyl-1H-pyrazol-4-yl)acetic acid methyl ester (160 mg, 0.33 mmol) in dioxan (2mL) is added an aqueous solutio of NaOH (1 M, 0.66 mL) and the mixture is stirred at 60°C for 1
-4816479
49/64 hour. The solvent is evaporated in vacuo, and the residue is suspended in water and treated with acetic acid (2 M). The precipitate is dryfreezed, dissolved in methanol and few dimethylformamide, and the product is precipitated with a few water, filtered and dried to yield 137 mg of the title compound (ESI mass spectrum: [M+Hf = 467; Rétention time HPLC: 0.89 min (method D)).
’H-NMR 400 MHz (DMSO-d6): δ [ppm] = 1.30 (t, 6H), 2.04 (s, 3H), 2.13 (s, 3H), 3.25 (s, 2H), 4.55 (q,4H), 5.16 (s, 2H), 6.97 (t, 1H), 7.17 (t, 1H), 7.29 (s, 1H), 7.47 (m, 1H), 7.62 (dd, 1H), 7.74 (d, 1H), 10.52 (s, 1H).
Compound 33: (1-(4-[(7-Chloro-3-methylbenzofuran-2-carbonyl)amino]benzyl)-3,5-dimethyl1H-pyrazol-4-yî)acetic acid (Coupling method C4):
a) To a solution of 7-chloro-3~methylbenzofuran-2-carboxylic acid (185 mg, 0.88 mmol) in 6 mL dichloromethane is added oxalyl chioride (0.123 mL, 1.14 mmol) and a drop of dimethylformamide. After stirring at room température for 1 hour the solvent is removed by évaporation in vacuo. The residue ts dissolved in dichloromethane (5 mL) and dropped to a solution of [1-(4-aminobenzyl)-3,5-dimethyl-1H-pyrazol-4-yl]acetic acid tert-butyl ester (428 mg, 1 mmol) in form ofthe toluenesulphonate saitand diisopropylethylamine (0.39 mL, 2.27 mmol) in dichloromethane (5 mL). After addition of 4-dimethylaminopyridine (11 mg, 0.09 mmol) the solution is stirred at room température for 12 hours. The solution is extracted twice with hydrochloric acid (1 M), twice with water, twice with an aqueous solution of NaOH (1 M) and twice with water. The organic layer is dried over MgSO4, filtered and the solvent is evaporated in vacuo to yield 426 mg of (1-(4-[(7-chloro-3-methyl-benzofuran-2carbonyl)amino]benzyl)-3,5-dimethyl-1H-pyrazol-4-yl)acetic acid tert-butyl ester (ESI mass spectrum: [M+Hf = 508; Rétention time HPLC: 1.60 min (method H)).
b) To a solution of (1-(4-[(7-chloro-3-methyl-benzofuran-2-carbonyl)amino]benzyl)-3,5dimethyl-1H-pyrazol-4-yl)acetic acid tert-butyl ester (426 mg, 0.84 mmol) in dichloromethane (10mL) is added trifluoroacetic acid (400 mL, 5.2 mmol) and the mixture is stirred at room température for 3 days. The solvent is evaporated in vacuo, and the residue is suspended in water and treated with diethylether and the precipitating product is filtered off to yield 174 mg of the title compound (ESI mass spectrum: [M+H]* = 452; Rétention time HPLC: 1.39 min (method J)).
-4916479
50/64 ’H-NMR 400 MHz (DMSO-d6): δ [ppm] = 2.07 (s, 3H), 2.14 (s, 3H), 2.57 (s, 3H), 3.29 (s, 2H), 5.17 (s, 2H), 7.12 (d, 2H), 7.38 (t, 1H), 7.62 (d, 1H), 7.74 (d, 2H), 7.76 (d, 1 H). 10.33 (s, 1H).
Compound 48: (1-(4-[(3-Ethyl-5-fluorobenzofuran-2-carbonyl)amino]benzyl)-3,5-dimethyl-1Hpyrazol-4-yl)-acetic acid (Coupling method C4)
a) To a solution of 3-ethyl-5-fluorobenzofuran-2-carboxylic acid (450 mg, 2.16 mmol) in 10 mL dichloromethane is added oxalyl chloride (0.335 mL, 3.1 mmol) and a drop dimethylformamide. After stirring at room température for 1 hour the solvent is removed by évaporation in vacuo. The residue is dissolved in dichloromethane (10 mL) and dropped to a solution of [1-(4“aminobenzyl)-3,5-dimethyl-1H-pyrazol-4-yl]acetic acid tert-butyl ester (1.054 g, 2.16 mmol) in the form of the 4-toluenesulphonate sait and diisopropylethylamine (1.3 mL, 7.57 mmol) in dichloromethane (5 mL). After addition of 4-dimethylaminopyridine (26.4 mg, 0.216 mmol) the solution is stirred at room température for 12 hours. The solvent is evaporated in vacuo, dissolved in 100 mL ethyl acetate, and the solution is extracted twice with water, once with hydrochloric acid (0.5 M), once with water, once with an aqueous solution of NaHCO3 (5% by weight) and once with water. The organic layer is dried over MgSO4, filtered and the solvent is evaporated in vacuo. The residue is purified by MPLC (dichloromethane/methanol 98:2) to yield 150 mg of 1-(4-[(3-ethyl-5-fluorobenzofuran-2carbonyl)aminoJbenzyl)-3,5-dimethyl-1H-pyrazol-4-yl)-acetic acid tert-butyl ester (ESI mass spectrum: [M+Hf = 506; Rétention time HPLC: 1.02 min (method D)).
b) To a solution of 1-(4-[(3-ethyl-5-fluorobenzofuran-2-carbonyl)amino]benzyl)-3,5-dimethyl1H-pyrazol-4-yl)-acetic acid tert-butyl ester (150 mg, 0.30 mmol) in acetonitrile (25mL) is added montmorillonite KSF (300 mg) and the mixture is refluxed for 6 hours. The mixture is diluted to 400 mL with acetonitrile, refluxed for 5 minutes and filtered. The solvent is evaporated in vacuo and the residue is washed with dimethylformamide (300 mL). After filtration the solvent is removed by évaporation and the residue is treated with acetone (200 mL). After trituration the precipitate is filtered off and washed with acetone to yield 112 mg of the title compound (ESI mass spectrum: [M+H]+ = 450; Rétention time HPLC: 0.85 min (method E)).
1H-NMR 400 MHz (DMSO-d6): δ [ppm] = 1.21 (t, 6H), 2.04 (s, 3H), 2.11 (s, 3H), 3.09 (q, 4H), 3.27 (s, 2H), 5.16 (s, 2H), 7.09 (d, 2H), 7.33 (t, 1 H), 7.67 (m, 2H), 7.76 (d, 2H), 10.39 (s, 1H).^<“
-5016479
51/64
Compounds 34 to 47 and 49 to 56 of table 1 below have likewise been prepared in analogy to coupling method C4 using suitable starting amines and carboxylic acids.
IV) BIOLOGICAL ASSAYS
The compounds of formula (la) and (lb) according to the invention were tested using the following biological test methods to détermine their ability to displace PGD2 from the CRTH2 receptor and for their ability to antagonise the functîonal effects of PGD2 at the CRTH2 receptor in a whole system.
PREPARATION OF HUMAN CRTH2 RECEPTOR MEMBRANES AND RADIOLIGAND BINDING ASSAY
The binding of CRTH2 antagonists is determined using membranes prepared from Chinese hamster ovary cells (CHO-K1 cells) transfected with the human CRTH2 receptor (CHO-K1hCRTH2 cells, Perkin Elmer, Cat No ES-561-C). To produce cell membranes the CHO-K1hCRTH2 cells are cultured in suspension in CHO SFMII medium supplemented with 400 pg/ml G418. The cells are harvested by centrifugation at 300 g for 10 min at room température. The cell pellet is resuspended in Phosphate Buffer Saline (PBS) including a protease inhibitor mix (Complété, Roche) and adjusted to a concentration of 10E7 cells/ml.
The CHO-K1-hCRTH2 cells are disrupted by nitrogen décomposition to obtain the membrane préparation. Cell débris is removed by centrifugation (500 g at 4°C, 30 min) and the supernatant is transferred into fresh tubes followed by a second centrifugation at 40000 g for 1 h at 4 °C to sédiment the membranes. The membranes are suspended in SPA incubation buffer (50mM Tris HCl, 10 mM MgCI21 150 mM NaCI, 1 mM EDTA, pH 7.4) without bovine sérum albumin, homogenized by passing through a single use needle (Terumo, 23Gx1), and stored in aliquots at -80 °C.
The CRTH2 receptor binding assay is performed in a scintillation proximity assay (SPA) format with the radioligand [3H]-PGD2 (Perkin Elmer, NET616000MC). CHO-K1-hCRTH2 cell membranes are again homogenized by passing through a single use needle (Terumo, 23Gx1) and diluted in SPA incubation buffer in suitable concentrations (0.5 -10 pg protein/well). The SPA assay is set up in 96 well microtiter plates (Perkin Elmer, CatNo. 6005040) in SPA incubation buffer with a final volume of 200 pl per well and final
-5116479
52/64 concentration of 50 mM Tris-HCI, 10 mM MgCI2, 150 mM NaCI, 1 mM EDTA pH 7,4, 0,1% bovine sérum albumin). The SPA assay mixture contains 60 pl of the membrane suspension, 80 μΙ of Wheat Germ Agglutinin coated PVT beads (GE Healthcare, RPNQ-0001, 0.3 mg/well), 40 μΙ of [3H]-PGD2 diluted in SPA buffer to a final concentration of 1 nM (50 000 dpm) and 20 μΙ of the test compound (dissolved in dimethylsulfoxid). The SPA assay mixture is incubated for 3 h at room température. Bound radioactivity is determined with a scintillation counter (Micro Beta Trilux, Wallac).
The binding of [3H]-PGD2 to CHO-K1-hCRTH2 cell membranes is determined in the absence (total binding, Bo) and presence (non-specific binding, NSB) of unlabelled PGD2 (1 μΜ, Cayman Chemical, Cat No 12010) or a reference CRTH2 antagonist (10 μΜ CAY10471, Cayman Chemical, Cat No 10006735).
Détermination of the affinity of a test compound is calculated by subtraction of the nonspecific binding (NSB) from the total binding (Bo) or the binding in the presence of the test compound (B) at a given compound concentration. The NSB value is set to 100% inhibition. The Bo-NSB value is set to 0% inhibition.
% inhibition values were obtained at a defined compound concentration, e.g. at 1 μΜ, % inhibition ofthe test compound was calculated by the formula 100-((B-NSB)*100/(BoNSB)). % inhibition values above 100% are founded by assay variance.
The dissociation constant K, was calculated by itérative fitting of experimental data obtained at several compound concentrations over a dose range from 0.1 to 30 000 nM using the law of mass action based program easy sys (Schittkowski, Num Math 68, 129-142 (1994)).
CRTH2 CAMP FUNCTIONAL ASSAY PROTOCOL
The assay is conducted in CHO-K1-hCRTH2 cells. Intracellular cAMP is generated by stimulating the cells with 10 μΜ Forskolin, an adenylate cyclase activator. PGD2 is added to activate the CRTH2 receptor which results in the atténuation of the forskolin-induced cAMP génération. Test compounds are tested for their ability to inhibit the PGD2-mediated atténuation of the Forskolin-induced cAMP génération in CHO-K1-hCRTH2 cells. CHO-K1-hCRTH2 cells are cultured in roller bottles in CHO SFMII medium supplemented with 400ug/ml G418. The cells are harvested by centrifugation at 300 g for 10 min at room
-5216479
53/64 température. The cell pellet is washed and suspended in PBS. The cells are adjusted to a final concentration of 4x10E6 cells/ ml.
Test compounds are diluted in dimethylsulfoxid and tested at several compound concentrations over a dose range from 0.1 to 3 000 nM.
The cAMP levels are determined by an AlphaScreen cAMP assay (Perkin Elmer CatNo. 6760625M) in 384 well optiplates (PerkinElmer, CatNo. 6007290) with a total assay volume of 50 μΙ. 10 μΙ of cells (40.000 cells per well) are incubated for 30 min at 37 °C with 10 μΙ of a stimulation mix containing a final concentration of 10μΜ Forskolin, 30 nM PGD2, 0.5 mM IBMX, 5 mM HEPES, IxHBSS buffer, 0.1% BSA, adjusted to pH 7.4, and the test compound at various concentrations. Thereafter, 30 μΙ of a lysis and détection mix is added containing SA donor beads, biotinylated cAMP, anti-cAMP acceptor beads, 0.3% Tweeen-20, 5 mM HEPES, 0.1% BSA, adjusted to pH 7.4. After 2 h incubation time the AlphaScreen signal is read on an AlphaQuest-HTS instrument. The IC50 values are calculated by using the Prism software.
OTHER CRTH2 FUNCTIONAL ASSAY PROTOCOLS
The ability of the tested compounds to antagonise the functional effects of PGD2 at the CRTH2 receptor may also be demonstrated by methodology known in the art, such as by a whole cell binding assay, a GTPgS assay, a BRET assay, an inositol phosphate accumulation assay, an CRTH2 cell surface expression assay, a Ca2+ influx assay, an ERK phosphorylation assay, an cell migration assay, an eosinophil shape change assay, a Th2 cell degranulation assay, or a basophil activation assay as described by Mathiesen et al., Mol Pharmacol. 2005, 68:393-402; Mimura et al., J. Pharmacol. Exp. Ther., 2005, 314:244-51; Sandham étal., Bioorg. Med. Chem. Lett., 2007,17:4347-50; Sandham Bioorg. Med. Chem. Lett, 2009,19:4794-8; Crosignani étal., J Med Chem, 2008, 51:2227-43; Royer étal., Eur J Clin Invest, 2008, 38:663-71; Boehme et al., Int Immunol, 2009, 21:621-32; Sugîmoto étal., Pharmacol. Exp. Ther., 2003, 305:347-52; Monneretetal., J Pharmacol Exp Ther, 2005, 312:627-34; Xue et al., J. Immunol., 2005,175:6531-6.
Cell lines for expressing the CRTH2 receptor include those naturally expressing the CRTH2 receptor, such as AML14.3D10 and NCI-H292 cells (Sawyer et al., Br. J. Pharmacol., 2002, 137:1163-72; Chiba et al., Int. Arch. Allergy. Immunol., 2007,143 Suppl 1:23-7), those induced to express the CRTH2 receptor by the addition of chemicals, such as HL-60 or AML14.3D10 cells treated with, for example, butyric acid (Sawyer et al., Br. J. Pharmacol., γ/
-5316479
54/64
2002, 137:1163-72) or a cell line engineered to express a recombinant CRTH2 receptor, such as L1.2, CHO, HEK-293, K562 or CEM cells (Liu et al., Bioorg. Med. Chem. Lett., 2009,19:6840-4; Sugimoto et al., Pharmacol Exp Ther, 2003, 305:347-52; Hata et al., Mol. Pharmacol., 2005, 67:640-7; Nagata et al., FEBS Lett, 1999, 459:195-9).
Finally, blood or tissue cells, for example human peripheral blood eosinophils, isolated using methods as described by Hansel étal., J. Immunol. Methods., 1991, 145,105-110, or human Th2 cells isolated and treated as described by Xue et al., J. Immunol., 2005,175:6531-6, or human basophils isolated and characterized as described by Monneret et al., J. Pharmacol, Exp. Ther., 2005, 312:627-34 can be utilized in such assays.
In particular, the compounds of the présent invention hâve activity in binding to the CRTH2 receptor in the aforementioned assays and inhibit the activation of CRTH2 by CRTH2 ligands. As used herein, activity is intended to mean a compound demonstrating an inhibition of 50% at 1 μΜ or higher in inhibition, or a K| value < 1 μΜ, when measured in the aforementioned assays. Such a resuit is indicative of the intrinsic activity of the compounds as inhibitor of CRTH2 receptor activity. Antagoniste activities of selected compounds are shown in tables 1 and 2 below.
Table 1: Compounds of formula la”
Cmpd “R3^ R1 = R2 Z Ryl; RyZ; R*3; Ry4; Ry5 MS [M+Hf Rétention Time Ki [nM]
1 F ch3 NH H; H; H; H; H 421 1.04 method G 0.8
2 Cl ch3 NH H; H; H; H; H 437 1.36 min method B 0.5
3 F ch3 nch2- CeH5 H; H; H; H; H 511 0.98 min method D 0.2
4 H ch3 O CH3; H; F; H; H 436 0.85 min method D 0.8
-5416479
55/64
Cmpd R1 = R2 Z Ry1; Ryi; Ry3; Ry4; Rys MS [M+H]+ Rétention Time Ki [nM]
5 Cl ch3 0 H; H; H; H; H 438 0.86 min method D 0.8
6 H ch3 0 H; H; OCH3; H; H 434 0.77 min method D 0.6
7 H ch3 0 CH3; H; H; H; F 436 0.86 min method D 1.3
8 H ch3 0 H; H; CI; H; H 438 0.84 min method D 0.3
9 H ch3 0 H; H; H; H; OCH3 434 0.78 min method D 1.2
10 H ch3 NH H; H; H; H; H 403 1.24 min method B 0.2
11 H ch3 NH H; H; H; F; H 421 0.76 min method D 0.2
12 H ch3 NH H; H; F; H; H 421 0.76 min method D 0.2
13 H ch3 nch3 CH3; H; H; H; H 431 0.81 min method D 63.9
14 H ch3 NH CH3; H; OCH3; H; H 447 0.79 min method D 0.1
15 H ch3 S H; H; H; H; H 420 1.32 min method B 0.3
16 H ch3 NCH3 H; H; H; H; H 417 1.29 min method K 0.8
17 H ch3 NC2H5 H; H; H; H; H 431 1.35 min method K 0.4
18 H ch3 nch2CeHs H; H; H; H; H 493 0.88 min method D < 0.1
19 H c2h5 0 H; H; Cl; H; H 466 1.43 min method K 0.2
20 H c2hs NH H; H; H; H; H 431 1.30 min method K 0.3
21 H c2h5 nch3 H; H; H; H; H 445 0.84 min method D 0.3
22 H c2h5 nc2h5 H; H; H; H; H 459 0.87 min method D 0.2
23 H C2H5 NH H; H; F; H; H 449 0.80 min method D 0.3
24 H c2h5 nch3 CH3; H; H; H; H 459 0.84 min method D 9.9
25 H c2h5 NH H; H; H; F; H 449 1.34 min method K 0.2
26 H ch3 0 H; H; H; H; H 404 1.28 min method B 0.8
27 F ch3 0 CH3; H; F; H; H 454 0.90 min method D 0.6
28 H ch3 N(CH2)2 -ch3 H; H; H; F; H 463 0.85 min method E 0.1
-5516479
56/64
Cmpd R3' R1 = R2 Z Ry1; RyZ; R*3; Ry4; RyS MS [M+H]* Rétention Time Ki [nM]
29 H ch3 nc2h5 H; H; H; F; H 449 0.84 min method E 0.1
30 F ch3 nch3 H; H; H; H; H 435 0.85 min method C 1.0
31 F ch3 nc2h5 H; H; H; H; H 449 0.85 min method C 0.2
32 F ch3 NC2Hs H; H; F; H; H 467 0.89 min method D 0.2
33 H ch3 0 CH3; H; H; H; Cl 452 1.39 min method J 2.2
34 H ch3 0 CH2CH3; H; H; H; F 450 1.38 min method L 0.3
35 H ch3 0 CH2CH3; H; F; H; F 468 1.43 min method L 0.3
36 H ch3 0 H; H; H; Cl; H 438 1.32 min method M 0.2
37 F ch3 0 CH3; H; H; H; Cl 470 1.46 min method J 2.2
38 F ch3 0 CH2CH3; H; H; H; F 468 0.88 min method E 0.6
39 F ch3 0 H; H; H; Cl; H 456 1.40 min method M 0.4
40 F ch3 nc2h5 H; F; H; H; H 467 0.87 min method E 0.4
41 F ch3 nc2h5 H; H; H; F; H 467 0.87 min method E 0.1
42 F ch3 N(CH2)zch3 H; H; H; F; H 481 0.89 min method E 0.1
43 F ch3 0 CH2OCH3; H; H; H; H 466 0.86 min method D 2.3
44 F ch3 o CH3; H; Cl; H; H 470 0.95 min method D 0.3
45 F ch3 o CH3; H; Br; H; H 514 0.96 min method D 0.3
46 F ch3 s CH3; H; F; H; H 470 0.88 min method D 4.5
47 F ch3 0 CH2CH3; H; F; H; H 468 0.94 min method D 0.9
48 H ch3 0 CH2CH3; H; F; H; H 450 0.85 min method E 1.2
49 F ch3 0 CH3; H; OCH3; H; H 466 1.30 min method F 0.1
50 F ch3 0 CH3; H; H; H; F 454 1.32 min method F 0.4
51 F ch3 nch3 H; H; F; H; H 453 1.28 min method F 0.4
52 F ch3 0 (CH2)2CH3; H; F; H; H 482 0.87 min method D 0.7
53 F ch3 N(CH2)3ch3 H; H; H; H; H 477 1.41 min method O 0.2
-5616479
57/64
Cmpd R3' R1 = R2 Z Ry1; RyZ; Ry3; Ry4; Ry5 MS [M+Hf Rétention Time Ki [nM]
54 F ch3 N(CH2)2ch3 H; H; F; H; H 481 0.92 min method D 0.2
55 F ch3 N(CH2)3ch3 H; H; F; H; H 495 0.91 min method E 0.1
56 F ch3 N(CH2)2ch3 H; H; H; H; H 463 0.87 min method E 0.2
Table 2: Compounds of formula Ib”
3
Cmpd R1,R2 R3 Z Ry1; Ryz; Ry3; Ry4; Ry5 MS [M+H]+ Rétention Time Ki [nM]
57 ch3 H O H; H; H; H; H 404 1.29 min method B 8.8
58 ch3 H NH H; H; H; H; H 403 1.14 min method B 37.2
59 ch3 H S H; H; H; H; H 420 1.32 min method B 3.8
:o*5JüfL 2013
-5716479

Claims (23)

1. Pyrazole compounds of formula (la) or (lb) and pharmaceutically acceptable salts thereof, wherein
Ra and Rb are independently selected hydrogen, hydroxy, halogen, CrCe-alkyl, CrC8 haloalkyl, CrCe-alkoxy, Ci-C6-haloalkoxy and C3-C8-cycloalkylt or Ra and Rb together with the carbon atom they are bound to form may form a carbonyl group, or Ra and Rb together with the carbon atom they are bound to form a 3to 8-membered ring, wherein said ring may contain 1 or 2 heteroatoms selected from O, N and S as ring member and wherein the ring members of said ring may optionally be independently substituted by hydroxy, halogen, CiC6-alkyl, Ci-C6-haloalkyl, CrCe-alkoxy, CrC6-haloalkoxy and C3-C8-cycloalkyl;
Rc and Rd are independently selected hydrogen, hydroxy, halogen, Ci-C6-alkyl, CrCehaloalkyl, CrCe-alkoxy, CrC6-haloalkoxy and C3-C8-cycloalkyl, or Rc and Rd together with the carbon atom they are bound to form may form a carbonyl group, or Rc and Rd together with the carbon atom they are bound to form a 3to 8-membered ring, wherein said ring may contain 1 or 2 heteroatoms selected from O, N and S as ring member and wherein the ring members of
-5816479
59/64 said ring may optionally be independently substituted by hydroxy, halogen, Cr
C0-alkyl, CrCe-haloalkyl, CrCe-alkoxy, CrC6-haloalkoxy and C3-C8-cycloalkyl;
Y1, Y2, Y3, Y4 and Y5 are independently selected from N and CRy, wherein each Ry is independently selected from H, hydroxy, halogen, cyano, nitro, SF5, C(O)NRfR9, Ci-C6-alkyl, hydroxy-CrCe-alkyl, Ct-Cg-alkoxy-CrCg-alkyl, C3- C8cycloalkyl, Cj-Cg-haloalkyl, CrC6-alkoxy, Ci-C6-alkoxy-CrC6-alkoxy, Ci-C0haloalkoxy, C3-CB-cycloalkoxy, C-i-Ce-alkylamino, di-Ci-C6-alkylamino, Ci-C0alkylsulfonyl, phenyl, phenoxy, 5- or 6-membered heterocyclyl and 5- or 6membered heterocyclyloxy, wherein Rf and R0 are independently from each other selected from H, CrC6-alkyl, CrC6-haloalkyl, C3-C0-cycloalkyl, C3-Cecycloalkenyl and 5- or 6-membered heterocyclyl or Rf and R3 together with the nitrogen atom to which they are bound form a cyclic amine, which may comprise a further heteroatom selected from O, N and S as a ring member;
Z is selected from O, S and NRZ, wherein Rz is H, Cr^-alkyl or benzyl;
R1 and R2 independently from each other are selected from H, halogen, CrCe-alkyl, C2-C6-alkenyl, C2-C0-alkynyl, CrC6-alkoxy, C1-C0-alkylthio, -NR'R9, C3-C0cycloalkyl, Cs-Ce-cycloalkyl-CrCe-alkyl, C3-C8-cycloalkyl-C2-C0-alkenyl, C3-C8cycloalkenyl, Cs-Ca-cycloalkenyl-CrCe-alkyl, C3-C8-cycloalkenyl-C2-C0-alkenyl, phenyl, phenyl-C^Ce-alkyl, phenyl-C2-C6-alkenyl, naphthyl, naphthyl-CrCsalkyl, naphthyl-C2-C0-alkenyl, heterocyclyl, heterocyclyl-Ci-C6-alkyl, and heterocyclyl-C2-C6-alkenyl, wherein the Ci-C6-alkyl, C2-C0-alkenyl and C2-C0-aikynyl moieties in the aforementioned radicals R1 and R2 are unsubstituted or carry at least one substituent selected from hydroxy, halogen, cyano, nitro, C1-C6-alkoxy, CrC6haloalkoxy, CrC6-alkylamino, di CrCg-alkylamino and CrC6-alkylsulfonyl and/or wherein two radicals bound to the same carbon atom of said CrC0-alkyl, C2C6-alkenyl and C2-C6-alkynyl moieties in the aforementioned radicals R1 and R2 together with said carbon atom may form a carbonyl group, and wherein
-5916479
60/64 the C3-C8-cycloalkyl, cycloalkenyl, phenyl, naphthyl and heterocyclyl moieties in the aforementioned radicals R1 and R2 are unsubstituted or carry at least one substituent selected from hydroxy, halogen, cyano, nitro, CrC6-alkyl, C3C8-cycloalkyl, Ci-C8-haloalkyl, CrC6-alkoxy, Ci-Ce-haloalkoxy, CrC6alkylamino, di-CrC6-alkylamino, CrCe-alkylsulfonyl, phenyl and 5- or 6membered hetaryl and/or wherein two radicals bound to the same carbon atom of said C3-C0-cycloalkyl, C3-Ce-cycloalkenyl and heterocyclyl moieties of the radicals R1 and R2 together with said carbon atom may form a carbonyl group, and wherein
Rf and R9 are independently from each other selected from H, Ci-C6-alkyl, Cr C6-haloalkyl, C3-C8-cycloalkyl, C3-C8-cycloalkenyl and heterocyclyl or
Rf and R9 together with the nitrogen atom to which they are bound form a cyclic amine, which may comprise a further heteroatom selected from O, N and S as a ring member;
n is an integer selected from 0, 1, 2 or 3; and
R3 if présent are selected independently from each other from halogen, ΟρΟβalkyl, Ci-C6-haloalkyl, Ci-C6-alkoxy, CrC6-haloalkoxy and C3-C8-cycloalkyl.
2. Pyrazole compounds of formula (la) or (lb) according to claim 1, wherein Ra and Rb are both hydrogen.
3. Pyrazole compounds of formula (la) or (lb) according to any of the preceding claims, wherein Rc and Rd are both hydrogen.
4. Pyrazole compounds of formula (la) or (lb) according to any of the preceding claims, wherein Y1 is CRy1 or N, wherein Ry1 has one of the meanings as defined for Ry in claim 1.
5. Pyrazole compounds of formula (la) or (lb) according to claim 4, wherein Y1 is CRy1.
-6016479
61/64
6. Pyrazole compounds of formula (la) or (lb) according to claim 5, wherein Ry1 is selected from H, CrCB-alkyl, CrC6-alkoxy-CrC6-alkyl and Ci-C6-haloalkyl.
7. Pyrazole compounds of formula (la) or (lb) according to any of the preceding claims, wherein Y2 is CR1'2, Y3 is CR*3, Y4 is CR*4 and Y5 is CR*5, wherein R*2, Ry3, Ry4 and R*5 independently from each other hâve one of the meanings as defined for Ry in claim 1.
8. Pyrazole compounds of formula (la) or (lb) according to claim 7, wherein Ry2, Ry3, Ry4 and R*5 are independently selected from H, halogen, CrCs-alkoxy, CrC6-alkoxy-Cr C6-alkoxy and CrC6-haloalkoxy.
9. Pyrazole compounds of formula (la) or (lb) according to any of the preceding claims, wherein Z is O.
10. Pyrazole compounds of formula (la) or (lb) according to any of claims 1 to 8, wherein ZisS.
11. Pyrazole compounds of formula (la) or (lb) according to any of claims 1 to 8, wherein Z is and NR1.
12. Pyrazole compounds of formula (la) or (lb) according to any of the preceding claims, wherein R1 and R2 independently from each other are selected from H, CrCB-alkyl, C3-CB-cycloalkyl, phenyl and naphthyl.
13. Pyrazole compounds of formula (la) or (lb) according to claim 12, wherein R1 and R2 independently from each other are selected from H, CrC4-alkyl, C3-C6-cycloalkyl and phenyl.
14. Pyrazole compounds of formula (la) or (lb) according to claim 13, wherein R1 and R2 are selected from CrC4-alkyl.
15. Pyrazole compounds of formula (la) or (lb) according to any of the preceding claims, wherein n is 0 or 1. ---6116479
62/64
16. Pyrazole compounds of formula (la) or (Ib) according to any of the preceding daims, wherein R3 if présent are independently selected from halogen.
17. Pyrazole compounds of formula (la) according to any of the preceding daims, wherein the pyrazole compounds are selected from compounds of formula (la’), wherein Z, R1, R2, R3, Ry1, R*2, Ry3, Ry4 and R75 hâve one of the meanings given in any of the preceding daims and n is 0 or 1.
18. Pyrazole compounds of formula (Ib) according to any of daims 1 to 16, wherein the pyrazole compounds are selected from compounds of general formula (Ib’), wherein Z, R1, R2, R3, Ry1, R*2, R*3, Ry4 and R16 hâve one of the meanings given in any of the preceding daims and n is 0 or 1.
19. Use of pyrazole compounds of formula (la) or (Ib) according to any of the preceding daims as médicaments.
20. Use of pyrazole compounds of formula (la) or (Ib) according to any of daims 1 to 18, for preparing a médicament for the treatment of diseases related to CRTH2 activity.
21. Use of pyrazole compounds of formula (la) or (Ib) according to any of daims 1 to 18 for preparing a médicament for the prévention and/or treatment of inflammatory,
-6216479
63/64 înfectious and immunoregulatory disorders, respiratory or gastrointestinal diseases or complaints, inflammatory diseases of the joints and allergie diseases of the nasopharynx, eyes, and skin.
5
22. Pharmaceutical formulations, containing one or more of the pyrazole compound of formula (la) and/or (Ib) according to any of ciaims 1 to 18.
23. Pharmaceutical formulations, containing one or more pyrazole compound of formula (la) and/or (Ib) according to any of ciaims 1 to 18 in combination with one or more io active substances selected from among betamimetics, anticholinergics, corticosteroids, PDE4 inhibitors, LTD4 antagonists, EGFR inhibitors, CCR3 antagonists, CCR5 antagonists, CCR9 antagonists, 5-LO inhibitors, histaminereceptor antagonists, SYK inhibitors and sulfonamides. uX
OA1201300282 2011-01-24 2012-01-20 Pyrazole compounds as CRTH2 antagonists. OA16479A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP11151876.7 2011-01-24

Publications (1)

Publication Number Publication Date
OA16479A true OA16479A (en) 2015-10-21

Family

ID=

Similar Documents

Publication Publication Date Title
CA2788224C (en) Pyrazole compounds as crth2 antagonists
AU2012210718B2 (en) Pyrazole compounds as CRTH2 antagonists
US9206164B2 (en) Pyrazole compounds as CRTH2 antagonists
OA16479A (en) Pyrazole compounds as CRTH2 antagonists.
NZ611695B2 (en) Pyrazole compounds as crth2 antagonists