WO2008075077A1 - Piperidine derivatives for the treatment of obesity - Google Patents

Piperidine derivatives for the treatment of obesity Download PDF

Info

Publication number
WO2008075077A1
WO2008075077A1 PCT/GB2007/004936 GB2007004936W WO2008075077A1 WO 2008075077 A1 WO2008075077 A1 WO 2008075077A1 GB 2007004936 W GB2007004936 W GB 2007004936W WO 2008075077 A1 WO2008075077 A1 WO 2008075077A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
optionally substituted
phenyl
piperidine
carbonyl
Prior art date
Application number
PCT/GB2007/004936
Other languages
French (fr)
Inventor
Roger John Butlin
Peter William Rodney Caulkett
Andrew Leach
Nicholas John Newcombe
Charles John O'donnell
James Matthew Wood
Original Assignee
Astrazeneca Ab
Astrazeneca Uk Limited
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 Astrazeneca Ab, Astrazeneca Uk Limited filed Critical Astrazeneca Ab
Publication of WO2008075077A1 publication Critical patent/WO2008075077A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/08Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms
    • C07D211/18Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D211/34Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms with hydrocarbon radicals, substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present invention relates to urethane derivatives, to processes for preparing such compounds, to their use as Fatty Acid Synthase inhibitors, to methods for their therapeutic use, particularly in the treatment of obesity and diabetes mellitus, and to pharmaceutical compositions containing them.
  • Obesity and diabetes are reaching epidemic proportions in the USA, EU, Japan and developing countries.
  • Obesity is the major driver of the co-morbidities of the metabolic syndrome, particularly type 2 diabetes. Since no effective pharmacotherapies for obesity are available to date and current diabetes therapies do not stop the progression of the disease, there is a huge unmet medical need.
  • Fatty Acid Synthase is a critical enzyme for endogenous lipogenesis and plays an important role in the modulation of key intermediates of lipid and carbohydrate cellular metabolism. FAS is highly expressed in the tissues with high metabolic activity
  • FAS inhibitor would cause beneficial metabolic effects in peripheral tissues.
  • inhibition of FAS in the hypothalamus may result in reduced food intake.
  • the non-specific irreversible FAS inhibitors cerulenin and C-75 have been reported in the literature to decrease brain levels of orexigenic neuropeptides and to decrease food intake.
  • the present invention provides a compound of formula I
  • R 1 represents 1) a C 1-6 alkyl group optionally substituted by one or two groups selected from A-S below and/or by one to five groups selected from T below: A) phenyl optionally substituted by one or more of the following i) halo; ii) cyano; iii) a Ci -4 alkoxy group optionally substituted by one or more halo iv) hydroxy; v) a C 1-4 alkyl group optionally substituted by one or more halo; vi) carbamoyl; vii) N-C 1- ⁇ alkylcarbamoyl; viii) N,N-diCi.6alkylcarbamoyl ; ix) carboxy; x) Ci -6 alkoxycarbonyl; xi) Ci- 6 alkylthio; xii) Ci-oalkylsulfinyl; xiii) Q.oalkyls
  • alkylcarbamoyl k
  • heteroaryl optionally substituted by one or more carboxy; fluoro; hydroxy; a C] -3 alkoxyo group optionally substituted on C2 or C3 by carboxy; a group ⁇ R c R d in which R c and R d are as defined above; or a group CONR e R f in which R e and R f are as defined above;
  • a C 3-1 ocycloalkyl group which may be monocyclic, bicyclic or tricyclic and optionally may be bridged and is optionally substituted by one or more carboxy; fluoro; hydroxy; a
  • SO 2 which is optionally fused to a benz ring and any ring is optionally substituted by0 a group A to T as defined above one or more of the following: hydroxy, oxo, carboxy, a
  • Ci -4 alkanoyl benzoyl, amino, G 1 . 3 alkylamino, (Ii(C 1-3 alkyl)amino or a Ci ⁇ alkyl optionally substituted by one or more hydroxy or C 1-6 alkoxy;
  • a C 2-6 alkenyl group optionally substituted by one or two groups selected from A to T above; 6) optionally substituted phenyl including optional fusion of the phenyl ring to a saturated or partially unsaturated 5 to 6 membered heterocyclic ring optionally containing one, two or three hetero atoms selected from oxygen, sulphur optionally in its oxidised forms of SO or SO 2 or nitrogen wherein the heterocyclic ring is optionally substituted by one or more of the following: a C ⁇ alkoxy group; a Ci -6 alkanoyl group;carboxy; a C 1-6 alkylsulfonyl group; a Ci- ⁇ alkoxycarbonyl group; carbamoyl; N-Ci- ⁇ alkylcarbamoyl; N, N-diQ.
  • ⁇ alkylcarbamoyl; hydroxy; oxo; a Ci ⁇ alkyl group (which is optionally substituted by one or more of the following: a Q- ⁇ alkoxy group, hydroxy or a group of formula ⁇ R c R d in which R° and R d are as defined above) and wherein the phenyl ring is optionally substituted by one or more of the groups i to xxix listed above or by a heteroaryl group optionally substituted by one or more groups i) to xxix) above or by an ureido group of formula R m R n N-C(O)-NH- in which R m and R n independently represent H, a C 1-6 alkyl group optionally substituted by a group, or R m and R n together with the nitrogen atom to which they are attached represent a saturated or partially unsaturated 4 to 8 membered heterocyclic ring optionally containing an additional sulphur including oxidised as SO or SO 2
  • R 2 represents H, halo, a Ci_ 3 alkyl group, trifluoromethoxy or cyano
  • R 3 represents H, halo, a Ci -3 alkyl group, trifluoromethoxy or cyano
  • R 4 represents i) H, ii) a C ⁇ alkyl group optionally substituted by one or more halo iii) a C 1- 3 alkoxy group optionally substituted by one or more halo iv) halo, v) nitro, vi) cyano, vii) a C 1-6 alkylS(O) y (O) z - wherein y is 0,1 or 2 and z is 0 except when y is 2 when z is 0 or 1 viii) a group CH 2 NR U R V in which R u and R v independently represent H; a C 1-3 alkylsulphonyl group, a C 1-3 alkanoyl group or a Ci_ 3 alkyl group
  • R 6 and R 6 independently represent H, halo, cyano, C 1-3 alkyl optionally substituted by one or more halo or C 1-3 alkoxy optionally substituted by one or more halo;
  • R 7 is H or OH.
  • R 7 is H.
  • R 1 represents 1) a C ⁇ alkyl group optionally substituted by one or more of the following: a) phenyl b) pyridyl or c) a Ci -3 alkoxy group 2) a C 3-7 cycloalkyl group or 3) phenyl ;
  • R 2 represents H, halo, a Ci -3 alkyl group; or trifluoromethoxy;
  • R 3 represents H, halo, a C 1-3 alkyl group; or trifluoromethoxy; provided that one of R 2 and R 3 is other than H;
  • R 4 represents cyano.
  • R 1 represents a Ci_ 6 alkyl group optionally substituted by 3-pyridyl or 4-pyridyl.
  • R 2 represents methyl and R 3 is H.
  • “Pharmaceutically acceptable salt”, where such salts are possible, includes both pharmaceutically acceptable acid and base addition salts.
  • a suitable pharmaceutically acceptable salt of a compound of formula I is, for example, an acid-addition salt of a compound of formula I which is sufficiently basic, for example an acid-addition salt with an inorganic or organic acid such as hydrochloric, hydrobromic, sulphuric, trifluoroacetic, citric or maleic acid; or, for example a base-addition salt of a compound of formula I which is sufficiently acidic, for example an alkali or alkaline earth metal salt such as a sodium, calcium or magnesium salt, or an ammonium salt, or a salt with an organic base such as methylamine, dimethylamine, trimethylamme, piperidine, morpholine or tris-(2-hydroxyethyl)amine.
  • a given chemical formula or name shall encompass all stereo and optical isomers and racemates thereof as well as mixtures in different proportions of the separate enantiomers, where such isomers and enantiomers exist, as well as pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates including solvates of the free compounds or solvates of a salt of the compound.
  • Isomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation.
  • the enantiomers may be isolated by separation of racemate for example by fractional crystallisation, resolution or HPLC.
  • the diastereomers may be isolated by separation of isomer mixtures for instance by fractional crystallisation, HPLC or flash chromatography.
  • stereoisomers may be made by chiral synthesis from chiral starting materials under conditions that will not cause racemisation or epimerisation, or by derivatisation, with a chiral reagent. All stereoisomers are included within the scope of the invention. AU tautomers, where possible, are included within the scope of the invention.
  • the present invention also encompasses compounds containing one or more isotopes for example 14 C, 11 C or 19 F and their use as isotopically labelled compounds for pharmacological and metabolic studies.
  • the present invention also encompasses prodrugs of a compound of formula I that is compounds which are converted into a compound of formula I in vivo. The following definitions shall apply throughout the specification and the appended claims.
  • C 3- iocycloalkyl group which may be monocyclic, bicyclic or tricyclic and optionally may be bridged” includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, bicyclo(2.2.1)heptyl, bicyclo(2.2.2)octyl, perhydroindanyl and adamantyl.
  • heteroaryl includes pyrrolyl, thienyl, furyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiadiazolyl, 1,3,4-oxadiazolyl, 1,2,4- oxadiazolyl, triazolyl, furazanyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, 1,3,5-triazinyl quinolyl, isoquinolyl, benzthienyl, benzofuranyl, benzofurazanyl, benzoxazolyl, benzimidazolyl, indolyl, benzthiazolyl, indazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, phthalazinyl, 1,5-naphth
  • heteroaryl including N-oxides includes heteroaryls as described immediately above and in addition N-oxides of such heteroaryls where such N-oxides are known to those skilled in the art to exist and are known to be stable at ambient conditions for example pyridine-N-oxides.
  • a carbon linked saturated or partially saturated 3 to 8 membered heterocyclic group containing one or more ⁇ , S, SO, SO 2 or O includes oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, 2,3-dihydro-l,3-thiazolyl, 1,3-thiazolidinyl, 1,3-oxazolidinyl, oxepanyl, azetidinyl, pyrrolinyl, pyrrolidinyl, morpholinyl, thiamorpholinyl (perhydro-l,4-thiazinyl), (8-oxa-3-azabicyclo[3.2.1]octyl), (7-oxa-3- azabicyclo[3.1.1 ]heptyl), perhydroazepinyl, perhydrooxazepinyl, tetrahydro- 1 ,4-thiazinyl, 1-oxoxo
  • alkyl denotes either a straight or branched alkyl group.
  • alkyl include methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, sec-butyl , t-butyl, pentyl, isopentyl, neopentyl, tert-pentyl, hexyl and isohexyl.
  • Preferred alkyl groups are methyl, ethyl, propyl, isopropyl, butyl and tertiary butyl.
  • alkoxy denotes a group O-alkyl, wherein alkyl is as defined above.
  • halogen shall mean fluorine, chlorine, bromine or iodine.
  • An is acetoxy.
  • Examples of “C ⁇ alkoxycarbonyl” include C] -4 alkoxycarbonyl, methoxycarbonyl, ethoxycarbonyl, n- and t-butoxycarbonyl.
  • Examples of “C 1-6 alkoxycarbonylamino” include methoxycarbonylamino, ethoxycarbonylamino, n- and ⁇ -butoxycarbonylamino.
  • Examples of “C ⁇ alkoxy” include methoxy, ethoxy and propoxy.
  • Examples of “C ⁇ alkanoylamino” include formamido, acetamido and propionylamino.
  • Examples of “C 1-6 alkylS(O) a wherein a is 0 to 2” include Ci -4 alkylsulphonyl, methylthio, ethylthio, methylsulphinyl, ethylsulphinyl, mesyl and ethylsulphonyl.
  • Examples of “C ⁇ alkylsulphonylamino” include methylsulphonylamino, ethylsulphonylamino and propylsulphonylamino.
  • Examples of "Ci -6 alkylsulphonyl-N-(C 1-6 alkyl)amino” include methylsulphonyl-N-methylamino, ethylsulphonyl-N-methylamino and propylsulphonyl-N-ethylamino.
  • Examples of "Ci-ealkanoyl” include C 1-4 alkanoyl, propionyl and acetyl. Examples of include methylamino and ethylamino.
  • Examples of "N,N-(Ci -6 alkyl) 2 amino include di-N-methylamino, di-(N-ethyl)amino and N-ethyl-N-methylamino.
  • Examples of “C 2-6 alkenyl” are vinyl, allyl and 1-propenyl.
  • Examples of “C 2-6 alkynyl” are ethynyl, 1-propynyl and 2-propynyl.
  • Examples of “N-(Ci -6 alkyl)sulphamoyl” are N-(methyl)sulphamoyl and N-(ethyl)sulphamoyl.
  • Examples of “N-(Ci- 6 alkyl) 2 Sulphamoyl” are N,N-(dimethyl)sulphamoyl and
  • N-(methyl)-N-(ethyl)sulphamoyl N-(methyl)-N-(ethyl)sulphamoyl.
  • ⁇ (C ⁇ ealky ⁇ carbamoyl are N-(Ci- 4 alkyl)carbamoyl, methylaminocarbonyl and ethylaminocarbonyl.
  • N,N-(Ci. 6 alkyl)2carbamoyl are N,N-(C ]-4 alkyl)carbamoyl, dimethylaminocarbonyl and methylethylaminocarbonyl.
  • C 3-8 Cy cloalkyl ring are cyclopropyl and cyclohexyl.
  • (heterocyclic group)C 1-6 alkyl include pyridylmethyl,
  • C 3-morpholinopropyl and 2-pyrimid-2-ylethyl examples include cyclopropylmethyl and 2-cyclohexylpropyl.
  • C 3- 8cycloalkylCi-6cycloalkyl examples include cyclopropylmethyl and 2-cyclohexylpropyl.
  • N-(Ci- 6 alkyl)sulphamoylamino are N-(methyl)sulphamoylamino andN-(ethyl)sulphamoylamino.
  • N-(C 1- 6alkyl) 2 sulphamoylamino are N,N-(dimethyl)sulphamoylamino and N-(methyl)-N-(ethyl)sulphamoylamino.
  • Ci-ealkylsulphonylaminocarbonyl examples include methylsulphonylaminocarbonyl, ethylsulphonylaminocarbonyl and propylsulphonylaminocarbonyl.
  • Specific compounds of the invention include one or more including any combination of the following compounds below labelled as List 1: 2-methylpropyl ⁇ -[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl- phenyl]carbamate; propan-2-yl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate benzyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate; phenyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate; benzyl N-[2-chloro-5-[4-(4-cyanophenyl)piperidine-l-carbonyl]phenyl]carbamate; benzyl N-[5-
  • a compound of the Formula I, or a pharmaceutically-acceptable salt thereof may be prepared by any process known to be applicable to the preparation of chemically-related compounds. Such processes, when used to prepare a compound of the formula I are provided as a further feature of the invention and are illustrated by the following representative process variants. Necessary starting materials may be obtained by standard procedures of organic chemistry. The preparation of such starting materials is described in conjunction with the following representative process variants and within the accompanying Examples. Alternatively necessary starting materials are obtainable by analogous procedures to those illustrated that are within the ordinary skill of an organic chemist.
  • a process for preparing a compound of formula I or a pharmaceutically acceptable salt thereof which process comprises : (a) reacting a compound of formula VI
  • X is a leaving group, for example halo, e.g. chloro, in the presence of a diluent, for example an organic liquid, e.g. dichloromethane, optionally in presence of a base, for example pyridine, at a temperature in the range of 0-150 0 C.
  • a diluent for example an organic liquid, e.g. dichloromethane
  • a base for example pyridine
  • X in which X represents a replaceable group, eg. Cl, Br, I, OMesyl, or OTriflyl in the presence of carbon monoxide and in the presence of a metal catalyst, eg. Pd or derivatives thereof, and in a solvent such as an alcohol, THF, toluene, or DMF, and in the temperature range 0 - 15O 0 C.
  • the carbon monoxide may be gaseous or in the form of a metal carbonyl, eg. Molybdenum hexacarbonyl.
  • Compounds of formula I may also be prepared by reacting a compound of formula VIII
  • Xl optionally in the presence of a coupling agent and optionally in the presence of a diluent for example a solvent at a temperature in the range of 0-15O 0 C.
  • Examples of coupling agents are Dichlorotriphenyl phosphorane (DCTPP), 1-ethyl-
  • HTBU N,N,N',N'-tetramethyluronium hexafluorophosphate
  • HATU O-(7-azabenzotriazol-l-yl)- N,N,N',N'-tetramethyluronium hexafluorophosphate
  • DTMM 4-(4,6-dimethoxy-l ,3,5- triazin-2-yl)-4-methylmorpholinium chloride
  • optional additives are: 1 -hydroxy benzotriazole (HOBt), 4- dimethylamino pyridine (DMAP), di- ⁇ o-propylethylamine (DIPEA), and triethylamine
  • TSA tetrahydrofuran
  • suitable solvents are: dimethyl formamide (DMF), chloroform, dichloromethane (DCM), and tetrahydrofuran (THF).
  • DMF dimethyl formamide
  • DCM dichloromethane
  • THF tetrahydrofuran
  • Compounds of formula I in which R 1 represents an optionally substituted pyridyl- s N-oxide may be prepared by reacting a compound of formula I in which R 1 represents an optionally substituted pyridyl with an oxidising agent for example urea hydrogen peroxide or 3-chloroperbenzoic acid, in the presence of a diluent for example dichloromethane or acetonitrile at a temperature in the range of 0-150°C.
  • an oxidising agent for example urea hydrogen peroxide or 3-chloroperbenzoic acid
  • compounds of formula I containing a sulphide group may be I 0 oxidised to SO or SO 2 for example by use of potassium peroxymonosulfate, nitriles may be reduce to aniinomethyl compounds, amines may be acylated or sulphonated to give amides or sulphonamides, respectively, activated heteroaryl halides may be hydrolysed to hydroxy groups, and esters may be hydrolysed to acids.
  • the compounds of the invention will normally be administered via the oral, parenteral, intravenous, intramuscular, subcutaneous or in other injectable ways, buccal, rectal, vaginal, transdermal and/or nasal route and/or via inhalation, in the form of
  • compositions comprising the active ingredient or a pharmaceutically acceptable addition salt, in a pharmaceutically acceptable dosage form.
  • the compositions may be administered at varying doses.
  • 3 o of humans are about 0.001-10 mg/kg body weight, preferably 0.01-1 mg/kg body weight.
  • Oral formulations are preferred particularly tablets or capsules which may be formulated by methods known to those skilled in the art to provide doses of the active compound in the range of 0.5mg to 500mg for example 1 mg, 3 mg, 5 mg, 10 mg, 25mg, 50mg, lOOmg and 250mg.
  • a pharmaceutical formulation comprising a compound of formula I, or pharmaceutically acceptable salt thereof, including the compound of the proviso, in admixture with pharmaceutically acceptable adjuvants, diluents and/or carriers.
  • the compounds of formula (I) are useful for the treatment of obesity or being overweight, (e.g., promotion of weight loss and maintenance of weight loss), prevention of weight gain (e.g., medication-induced or subsequent to cessation of smoking), for modulation of appetite and/or satiety, eating disorders (e.g. binge eating, bulimia and compulsive eating), dyslipidaemia and the treatment of type 2 diabetes mellitus.
  • the present compounds of formula (I) are useful for the prophylaxis and/or treatment of clinical conditions associated with inherent or induced reduced sensitivity to insulin (insulin resistance) and associated metabolic disorders (also known as the metabolic syndrome). These clinical conditions will include, but will not be limited to, general obesity, abdominal obesity, arterial hypertension, hyperinsulinaemia, hyperglycaemia, type 2 diabetes and the dyslipidaemia characteristically appearing with insulin resistance.
  • This dyslipidaemia also known as the atherogenic lipoprotein profile, is characterised by moderately elevated non-esterified fatty acids, elevated very low density lipoprotein (VLDL) triglyceride rich particles, high Apo B levels, low high density lipoprotein (HDL) levels associated with low apoAI particle levels and high Apo B levels in the presence of small, dense, low density lipoproteins (LDL) particles, phenotype B.
  • VLDL very low density lipoprotein
  • HDL low high density lipoprotein
  • LDL low density lipoprotein
  • the compounds of the present invention are expected to be useful in treating patients with combined or mixed hyperlipidemias or various degrees of hypertriglyceridemias and postprandial dyslipidemia with or without other manifestations of the metabolic syndrome.
  • Treatment with the present compounds is expected to lower the cardiovascular morbidity and mortality associated with atherosclerosis due to their antidyslipidaemic as well as anti-inflammatory properties.
  • the cardiovascular disease conditions include macro- angiopathies of various internal organs causing myocardial infarction, congestive heart failure, cerebrovascular disease and peripheral arterial insufficiency of the lower extremities.
  • the compounds of formula I are also expected to prevent or delay the development of type 2 diabetes from the metabolic syndrome and diabetes of pregnancy. Therefore the development of long-term complications associated with chronic hyperglycaemia in diabetes mellitus, such as the micro-angiopathies causing renal disease, retinal damage and peripheral vascular disease of the lower limbs, is expected to be delayed.
  • the compounds may be useful in treatment of various conditions outside the cardiovascular system whether or not associated with insulin resistance, like polycystic ovarian syndrome, obesity, cancer and states of inflammatory disease including neurodegenerative disorders such as mild cognitive impairment, Alzheimer's disease, Parkinson's disease and multiple sclerosis.
  • the compounds of formula I may also be useful in the treatment of metabolic syndrome and Prader-Willi syndrome.
  • the present invention provides a compound of formula I as previously defined for use as a medicament. In a further aspect the present invention provides the use of a compound of formula
  • I in the preparation of a medicament for the treatment or prophylaxis of obesity or being overweight (e.g., promotion of weight loss and maintenance of weight loss), prevention of weight gain (e.g., medication-induced or subsequent to cessation of smoking), for modulation of appetite and/or satiety, eating disorders (e.g. binge eating, bulimia and compulsive eating) and for the treatment or prophylaxis of dyslipidaemia and for the treatment or prophylaxis of type 2 diabetes mellitus.
  • obesity disorders e.g. binge eating, bulimia and compulsive eating
  • the present invention provides a method of treating obesity or being overweight, (e.g., promotion of weight loss and maintenance of weight loss), prevention of weight gain (e.g., medication-induced or subsequent to cessation of smoking), for modulation of appetite and/or satiety, eating disorders (e.g. binge eating, bulimia and compulsive eating) dyslipidaemia and type 2 diabetes mellitus comprising administering a pharmacologically effective amount of a compound of formula I, including the compound of the proviso, to a patient in need thereof.
  • obesity or being overweight e.g., promotion of weight loss and maintenance of weight loss
  • prevention of weight gain e.g., medication-induced or subsequent to cessation of smoking
  • eating disorders e.g. binge eating, bulimia and compulsive eating
  • type 2 diabetes mellitus e.g. binge eating, bulimia and compulsive eating
  • administering a pharmacologically effective amount of a compound of formula I, including
  • Combination Therapy may be combined with another therapeutic agent that is useful in the treatment of obesity such as other anti-obesity drugs, that affect energy expenditure, glycolysis, gluconeogenesis, glucogenolysis, lipolysis, lipogenesis, fat absorption, fat storage, fat excretion, hunger and/or satiety and/or craving mechanisms, appetite/motivation, food intake, or G-I motility.
  • another therapeutic agent that is useful in the treatment of obesity
  • anti-obesity drugs that affect energy expenditure, glycolysis, gluconeogenesis, glucogenolysis, lipolysis, lipogenesis, fat absorption, fat storage, fat excretion, hunger and/or satiety and/or craving mechanisms, appetite/motivation, food intake, or G-I motility.
  • the compounds of the invention may further be combined with another therapeutic agent that is useful in the treatment of disorders associated with obesity such as hypertension, hyperlipidaemias, dyslipidaemias, diabetes, sleep apnea, asthma, heart disorders, atherosclerosis, macro and micro vascular diseases, liver steatosis, cancer, joint disorders, and gallbladder disorders.
  • a compound of the present invention may be used in combination with a another therapeutic agent that lowers blood pressure or that decreases the ratio of LDL:HDL or an agent that causes a decrease in circulating levels of LDL-cholesterol.
  • the compounds of the invention may also be combined with therapeutic agents used to treat complications related to microangiopathies.
  • the compounds of the invention may be used alongside other therapies for the treatment of obesity and its associated complications the metabolic syndrome and type 2 diabetes, these include biguanide drugs, insulin (synthetic insulin analogues) and oral antihyperglycemics (these are divided into prandial glucose regulators and alpha- glucosidase inhibitors).
  • the compound of formula I, or a pharmaceutically acceptable salt thereof may be administered in association with a PPAR modulating agent.
  • PPAR modulating agents include but are not limited to a PPAR alpha and/or gamma agonist, or pharmaceutically acceptable salts, solvates, solvates of such salts or prodrugs thereof. Suitable PPAR alpha and/or gamma agonists, pharmaceutically acceptable salts, solvates, solvates of such salts or prodrugs thereof are well known in the art.
  • the combination of the invention may be used in conjunction with a sulfonylurea.
  • the present invention also includes a compound of the present invention in combination with a cholesterol-lowering agent.
  • the cholesterol-lowering agents referred to in this application include but are not limited to inhibitors of HMG-CoA reductase (3- hydroxy-3-methylglutaryl coenzyme A reductase).
  • HMG-CoA reductase inhibitor is a statin.
  • cholesterol-lowering agent also includes chemical modifications of the HMG-CoA reductase inhibitors, such as esters, prodrugs and metabolites, whether active or inactive.
  • the present invention also includes a compound of the present invention in combination with an inhibitor of the ileal bile acid transport system (IBAT inhibitor).
  • IBAT inhibitor an inhibitor of the ileal bile acid transport system
  • the present invention also includes a compound of the present invention in combination with a bile acid binding resin.
  • the present invention also includes a compound of the present invention in combination with a bile acid sequestering agent, for example colestipol or cholestyramine or cholestagel.
  • a combination treatment comprising the administration of an effective amount of a compound of the formula I, or a pharmaceutically acceptable salt thereof, optionally together with a pharmaceutically acceptable diluent or carrier, with the simultaneous, sequential or separate administration one or more of the following agents selected from: a CETP (cholesteryl ester transfer protein) inhibitor; a cholesterol absorption antagonist; a MTP (microsomal transfer protein) inhibitor ; a nicotinic acid derivative, including slow release and combination products; a phytosterol compound ; probucol; an anti-coagulant; an omega-3 fatty acid ; another anti-obesity compound for example sibutramine, phentermine, orlistat, bupropion, ephedrine, thyroxine; an aldose reductase inhibitor;
  • NaSSA an antipsychotic agent for example olanzapine and clozapine; a serotonin receptor modulator; a leptin/leptin receptor modulator; a CBl receptor modulator for example an inverse agonist or an antagonist; a GLK receptor modulator; a DPP-IV inhibitor; a cholesterol absorption inhibitor; a GLP-I agonist; an SGLT-2 inhibitor; a DGATl inhibitor; a DGAT2 inhibitor; a DGAT2 inhibitor anti-sense oligonucleotide; a ghrelin antibody; a ghrelin antagonist; an l l ⁇ HSD-I inhibitor; an UCP- 1,2 or 3 activator; or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, optionally together with a pharmaceutically acceptable diluent or carrier to a warmblooded animal, such as man in need of such therapeutic treatment.
  • a combination treatment comprising the administration of an effective amount of a compound of the formula I, or a pharmaceutically acceptable salt thereof, optionally together with a pharmaceutically acceptable diluent or carrier, with the simultaneous, sequential or separate administration of very low calorie diets (VLCD) or low-calorie diets (LCD).
  • VLCD very low calorie diets
  • LCD low-calorie diets
  • a method for the treatment of obesity and its associated complications in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof in simultaneous, sequential or separate administration with an effective amount of a compound from one of the other classes of compounds described in this combination section, or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof.
  • a method of treating hyperlipidemic conditions in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof in simultaneous, sequential or separate administration with an effective amount of a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof.
  • a pharmaceutical composition which comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, in association with a pharmaceutically acceptable diluent or carrier.
  • kits comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof.
  • kits comprising: a) a compound of formula I, or a pharmaceutically acceptable salt thereof, in a first unit dosage form; b) a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof; in a second unit dosage form; and c) container means for containing said first and second dosage forms.
  • kits comprising: a) a compound of formula I, or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable diluent or carrier, in a first unit dosage form; b) a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, in a second unit dosage form; and c) container means for containing said first and second dosage forms.
  • a compound of the formula I or a pharmaceutically acceptable salt thereof, and one of the other compounds described in this combination section, or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, in the manufacture of a medicament for use in the the treatment of obesity and its associated complications in a warm-blooded animal, such as man.
  • a compound of the formula I or a pharmaceutically acceptable salt thereof, and one of the other compounds described in this combination section, or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, in the manufacture of a medicament for use in the treatment of hyperlipidaemic conditions in a warm-blooded animal, such as man.
  • a combination treatment comprising the administration of an effective amount of a compound of the formula I, or a pharmaceutically acceptable salt thereof, optionally together with a pharmaceutically acceptable diluent or carrier, with the simultaneous, sequential or separate administration of an effective amount of one of the other compounds described in this combination section, or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, optionally together with a pharmaceutically acceptable diluent or carrier to a warm-blooded animal, such as man in need of such therapeutic treatment.
  • a compound of the invention may also be combined with therapeutic agents that are useful in the treatment of disorders or conditions associated with obesity (such as type II diabetes, metabolic syndrome, dyslipidemia, impaired glucose tolerance, hypertension, coronary heart disease, non-alcoholic steatohepatitis, osteoarthritis and some cancers) and psychiatric and neurological conditions.
  • obesity such as type II diabetes, metabolic syndrome, dyslipidemia, impaired glucose tolerance, hypertension, coronary heart disease, non-alcoholic steatohepatitis, osteoarthritis and some cancers
  • psychiatric and neurological conditions such as type II diabetes, metabolic syndrome, dyslipidemia, impaired glucose tolerance, hypertension, coronary heart disease, non-alcoholic steatohepatitis, osteoarthritis and some cancers.
  • a patient may be identified by, for example, measuring body mass index (BMI), which is calculated by dividing weight in kilograms by height in metres squared, and comparing the result with the definitions.
  • BMI body mass index
  • the compounds of the invention may also be useful as anti-cell-proliferation (such as anti-cancer) agents and are therefore useful in methods of treatment of the human or animal body.
  • Such properties are expected to be of value in the treatment of disease states associated with cell cycle and cell proliferation such as cancers (solid tumors and leukemias), fibroproliferative and differentiative disorders, psoriasis, rheumatoid arthritis, Kaposi's sarcoma, haemangioma, acute and chronic nephropathies, atheroma, atherosclerosis, arterial restenosis, autoimmune diseases, acute and chronic inflammation, bone diseases and ocular diseases with retinal vessel proliferation.
  • cancers solid tumors and leukemias
  • fibroproliferative and differentiative disorders psoriasis, rheumatoid arthritis, Kaposi's sarcoma, haemangioma, acute and chronic nephropathies, atheroma, atherosclerosis, arterial restenosis, autoimmune diseases, acute and chronic inflammation, bone diseases and ocular diseases with retinal vessel proliferation.
  • anti-cancer treatment may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional surgery or radiotherapy or chemotherapy.
  • chemotherapy may include one or more of the following categories of anti-tumour agents:
  • antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology such as alkylating agents (for example cis-platin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan and nitrosoureas); antimetabolites (for example antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and
  • cytostatic agents such as antioestrogens (for example tamoxifen, toremifene, raloxifene, droloxifene and iodoxyfene), oestrogen receptor down regulators (for example fulvestrant), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5 ⁇ -reductase such as finasteride; (iii) agents which inhibit cancer cell invasion (for example metalloproteinase inhibitors like marimastat and inhibitors of urokinase plasminogen activator receptor function
  • antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, (for example the anti-vascular endothelial cell growth factor antibody bevacizumab [AvastinTM], compounds such as those disclosed in International Patent Applications WO 97/22596, WO 97/30035, WO 97/32856 and WO 98/13354) and compounds that work by other mechanisms (for example linomide, inhibitors of integrin ⁇ v ⁇ 3 function and angiostatin);
  • vascular endothelial growth factor for example the anti-vascular endothelial cell growth factor antibody bevacizumab [AvastinTM]
  • vastinTM anti-vascular endothelial cell growth factor antibody bevacizumab
  • compounds that work by other mechanisms for example linomide, inhibitors of integrin ⁇ v ⁇ 3 function and angiostatin
  • vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529, WO 00/41669, WO 01/92224, WO 02/04434 and WO 02/08213;
  • antisense therapies for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
  • gene therapy approaches including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCAl or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy; and
  • immunotherapy approaches including for example ex-vivo and in- vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies.
  • cytokines such as interleukin 2, interleukin 4 or granulocyte-m
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment.
  • Such combination products employ the compounds of this invention within the dosage range described hereinbefore and the other pharmaceutically-active agent within its approved dosage range.
  • the compounds of the present invention may also be useful as anti-infective agents or as anti-bacterial agents.
  • the compounds of the present invention may also be useful as in decreasing sebum production following topical application.
  • Pharmacological Activity may also be useful as in decreasing sebum production following topical application.
  • the compounds of the present invention are Fatty Acid Synthase inhibitors.
  • the activity of the compounds of the invention was demonstrated using the following assay. Human and Rat FAS Enzyme Assay.
  • Fatty acid synthase is an enzyme complex that harbours seven enzymatic activities catalysing the reductive synthesis of long chain fatty acids from acetyl CoA and malonyl CoA to palmitate.
  • NADPH is consumed forming NADP. Since NADPH is fluorescent but not NADP the reaction can be measured by analysing the decrease in fluorescence.
  • Fatty acid synthase Human or rat enzyme (0.4 ⁇ g, produced in house), dissolved in 2OmM Tris/HCl pH 7.5, 5mM BOG 5 ImM TCEP,10% glycerol,lmM EDTA,150mM NaCl, was then added to the plate in a volume of lO ⁇ l. Enzyme was added to all but the last two columns of the plate, to which, lO ⁇ l of assay buffer was added (0.1M Tris ph7.5, O.lmM EDTA, ImM glutathione, 0.05%BSA) to provide a no enzyme assay control.
  • assay buffer 0.1M Tris ph7.5, O.lmM EDTA, ImM glutathione, 0.05%BSA
  • the compounds of the present invention were found to inhibit the activation of Fatty Acid Synthase with IC50S in a range of about 0.00 l ⁇ M to about 30 ⁇ M in the above assay.
  • the examples of the present invention inhibited the activition of Fatty Acid Synthase with IC 5 oS in a range of about 0.00 l ⁇ M to about 5.0 ⁇ M.
  • the compounds inhibit the activation of Fatty Acid Synthase with IC 50S in a range of about 0.00 l ⁇ M to about O.l ⁇ M.
  • the compound of Example 1 had an ICs 0 Of 0.4 ⁇ M, the compound of Example 3 had an IC5 0 of 0.4 ⁇ M and the compound of Example 7 had an IC5 0 of 1.7 ⁇ M.
  • temperatures are given in degrees Celsius ( 0 C); operations were carried out at room or ambient temperature, that is, at a temperature in the range of 18-25 0 C, unless otherwiseo stated;
  • chromatography means flash chromatography on silica gel; thin layer chromatographys (TLC) was carried out on silica gel plates;
  • NMR data when given, NMR data is in the form of delta values for major diagnostic protons, given in parts per million (ppm) relative to tetramethylsilane (TMS) as an internal standard5 when the solvent is CDCl 3 (when the solvent is d 6- -DMSO, it locks on to the 2.49 DMSO peak), determined at 300 MHz unless otherwise indicated; the following abbreviations have been used: s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; b, broad;
  • Triphosgene (62 mg, 0.21 mmol) was added to a solution of 4-[l-(3-amino-4-methyl- benzoyl)-4-piperidyl]benzonitrile (Intermediate A, 167 mg, 0.52 mmol ) and DIPEA (180 ⁇ L) in anhydrous THF (8 mL).
  • the reaction mixture was purged with nitrogen and stirred at ambient temperature for 3 1 A hrs.
  • the reaction mixture was filtered and the filtrate added to a reaction tubes containing 2-propanol (1 mL, large excess). The reaction mixture was then heated with stirring at 65°C for 3hrs, and then concentrated in vacuo.
  • solvents examples include THF, DCM, other; examples of bases are TEA, DIPEA and pyridine, and the reactions may be performed at temperatures between O 0 C and the boiling point of the solvent.
  • Step 1 A solution of 3-nitro-4-(trifluoromethoxy)benzoic acid (Step 1) (3.51 g, 14 mmol) in MeOH (150 mL) was hydrogenated at ambient temperature and pressure in the presence of 10% palladium on charcoal catalyst (500 mg). The catalyst was removed by filtration and washed through with more MeOH. The filtrate and washings were combined and evaporated to give the title compound as a pale cream solid (2.9 g), 1 H NMR (300.073 MHz, dmso) ⁇ 5.60 (br s, 2H),7.07 - 7.23 (m, 2H),7.39 - 7.46 (m, 1H),12.O2 - 13.40 (br s, IH), m/z 220 (M-H)-. Step 3: 4- [ 1 - [3 -amino-4-(trifluoromethoxy)benzoyl] -4-piperidyl]benzonitrile
  • Step 2 A mixture of 3-amino-4-trifluoromethoxy benzoic acid (Step 2) (2.8 g, 12.66 mmol), 4-(4'- cyanophenyl)piperidine (2.36 g, 12.66 mmol, 1 eq), N-(3-Dimethylaminopropyi)-N'- ethylcarbodiimide hydrochloride (EDAC) (2.67 g, 13.93 mmol, 1.1 eq) and DMAP (155 mg, 1.27 mmol, 0.1 eq) in DMF (30 mL) was stirred at room temperature for 2 hrs.
  • EDAC N-(3-Dimethylaminopropyi)-N'- ethylcarbodiimide hydrochloride
  • DMAP 155 mg, 1.27 mmol, 0.1 eq
  • Step 2 4-[ 1 -(5-amino-2-methyl-benzoyi)-4-piperidyi]benzonitrile
  • Step 1 starting from 2,4-dimethyl-5-nitro-benzoic acid and 4-(4'-cyanophenyl)piperidine;
  • Step 2 4- [ 1 -(5 -amino-2,4-dimethyl-benzoyl)-4-piperidyl]benzonitrile
  • Step 1 A suspension of methyl 4-[l-(4-methyl-3-nitro-benzoyl)-4-pi ⁇ eridyl]benzoate (Step 1) (5.68 g, 14.9 mmol) in MeOH (57 mL) was treated with aqueous sodium hydroxide solution (19 mL of 2M, 37.1 mmol, 2 eq.) and the reaction mixture stirred at 5O 0 C for two hrs. More MeOH (25 mL) was added and stirring for continued for one hr. The reaction mixture was cooled and treated with 2M aqueous hydrochloric acid to ⁇ pH5, diluted with EtOAc, and the organic layer separated. The aqueous portion was shaken with more EtOAc and the organic layer again separated.
  • reaction mixture was washed with water and the phases separated. The organic portion was concentrated to a brown solid.
  • the crude product was re-dissolved in DCM and the solution washed sequentially with water, citric acid solution (IM in water) and saturated sodium bicarbonate solution.
  • Step 3 A solution of N,N-dimethyl-4-[l-(4-methyl-3-nitro-benzoyl)-4-piperidyl] benzamide (Step 3) (1.74g) in MeOH (35 tnL) was treated with palladium-on-charcoal catalyst (122 mg of 10% Pd/C). The reaction mixture was stirred in an atmosphere of hydrogen at ambient temperature and pressure for 2 hours.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Child & Adolescent Psychology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A compound of formula (I), or a pharmaceutically acceptable salt thereof, processes for preparing such compounds, their use as Fatty Acid Synthase inhibitors, methods for their therapeutic use, particularly in the treatment of obesity and diabetes mellitus, and pharmaceutical compositions containing them.

Description

PIPERIDINE DERIVATIVES FOR THE TREATMENT OF OBESITY
Field of invention
The present invention relates to urethane derivatives, to processes for preparing such compounds, to their use as Fatty Acid Synthase inhibitors, to methods for their therapeutic use, particularly in the treatment of obesity and diabetes mellitus, and to pharmaceutical compositions containing them.
Background of the invention
Obesity and diabetes are reaching epidemic proportions in the USA, EU, Japan and developing countries. Obesity is the major driver of the co-morbidities of the metabolic syndrome, particularly type 2 diabetes. Since no effective pharmacotherapies for obesity are available to date and current diabetes therapies do not stop the progression of the disease, there is a huge unmet medical need.
Fatty Acid Synthase (FAS) is a critical enzyme for endogenous lipogenesis and plays an important role in the modulation of key intermediates of lipid and carbohydrate cellular metabolism. FAS is highly expressed in the tissues with high metabolic activity
(for example liver, adipose tissue and brain) and there are good reasons to believe that a
FAS inhibitor would cause beneficial metabolic effects in peripheral tissues. In addition, inhibition of FAS in the hypothalamus may result in reduced food intake. The non-specific irreversible FAS inhibitors cerulenin and C-75 have been reported in the literature to decrease brain levels of orexigenic neuropeptides and to decrease food intake.
Therefore there is a need for an effective FAS inhibitor to treat obesity and diabetes.
Description of the invention
The present invention provides a compound of formula I
Figure imgf000002_0001
or a pharmaceutically acceptable salt thereof, in which R1 represents 1) a C1-6alkyl group optionally substituted by one or two groups selected from A-S below and/or by one to five groups selected from T below: A) phenyl optionally substituted by one or more of the following i) halo; ii) cyano; iii) a Ci-4alkoxy group optionally substituted by one or more halo iv) hydroxy; v) a C1-4alkyl group optionally substituted by one or more halo; vi) carbamoyl; vii) N-C1- βalkylcarbamoyl; viii) N,N-diCi.6alkylcarbamoyl ; ix) carboxy; x) Ci-6 alkoxycarbonyl; xi) Ci-6alkylthio; xii) Ci-oalkylsulfinyl; xiii) Q.oalkylsulfonyl; xiv) Ci-βalkylsulfonyloxy; xv) sulphamoyl; xvi) N-C^alkylsulphamoyl; xvii) N, N-diCi-βalkylsulphamoyl; xviii) benzyl xix) benzyloxy; xx) heteroaryl; xxi) heteroaryloxy; xxii) phenyl xxiii) phenoxy xxiv) phenylsulphamoyl; xxv) heteroarylsulphamoyl; xxvi) a carbon linked saturated or partially unsaturated 4 to 8 membered heterocyclic group as defined in c) below; xxvii) phenylsulfonyl ; xxviii) heteroarylsulfonyl; xxix) a group of formula ΝRcRd in which Rc and Rd independently represent: a) H; b) C1-6alkanoyl; c) a carbon linked saturated or partially unsaturated 4 to 8 membered heterocyclic group containing one or more N, S or O, wherein the S may be in its oxidised form of SO or SO2 , which is optionally fused to a benz ring and any ring is optionally substituted by one or more of the following: hydroxy, oxo, carboxy, a Ci^alkoxy group optionally substituted by one or more hydroxy or C^alkoxy, C1-4alkanoyl, benzoyl, amino, Ci-3alkylamino, di(C1-3 alkyl)amino or a C1-6alkyl optionally substituted by one or more hydroxy or Ci-βalkoxy; d) a d-βalkyl group optionally substituted by one or more of the following: hydroxy; carboxy; a Ci_6alkoxycarbonyl group; a Ci-βalkoxy group; heteroaryl; a group of formula NReRf in which Re and Rf independently represent H; a C^alkanoyl group; a C1- βalkylsulphonyl group; a C1-6alkoxycarbonyl group; a Cμδalkyl group optionally substituted by one or more hydroxy or Ci-βalkoxy , or Re and Rf together with the nitrogen atom to which they are attached represent a saturated or partially unsaturated 4 to 8 membered heterocyclic ring optionally containing an additional sulphur including oxidised as SO or SO2 , oxygen or nitrogen and/or optionally fused to a benz ring and any ring is optionally substituted by one or more of the following: a Ci-6alkoxy group; carboxy; a Ci- 6alkylsulfonyl group; Ci-4alkanoyl; benzoyl; hydroxy; oxo; carboxy; or a
Figure imgf000003_0001
group optionally substituted by one or more hydroxy or by one or more C^alkoxy or by one or more carboxy; e) Rc and Rd together with the nitrogen atom to which they are attached represent a saturated or partially unsaturated 4 to 8 membered heterocyclic ring optionally containing an additional oxygen, sulphur, SO, SO2 or nitrogen and/or optionally fused to a benz ring and/or optionally substituted by one or more of the following: a C1-6alkoxy group; C1- 4alkanoylgroup; benzoyl; a d^alkoxycarbonyl group; a Ci-βalkylsulfonyl group; carbamoyl; N-Ci-6alkylcarbamoyl; N, N-diC^ealkylcarbamoyl; hydroxy; oxo; carboxy; a Ci-βalkyl group (which is optionally substituted by one or more of the following: a C1.o βalkoxy group, hydroxy or a group of formula ΝReRf in which Re and Rf are as defined above) or a group of formula NReRf in which Re and Rf are as defined above; f) a Ci-βalkylsulphonyl group; g) phenylsulfonyl; h) heteroarylsulfonyl; s i) benzoyl; j) phenyl optionally substituted by one or more of the following: halo; C1-3alkyl; C1- 3alkoxy; a C1-6alkanoylamino group; carbamoyl; N-Q-βalkylcarbamoyl; N,N-diC]. δalkylcarbamoyl; k) heteroaryl optionally substituted by one or more carboxy; fluoro; hydroxy; a C]-3alkoxyo group optionally substituted on C2 or C3 by carboxy; a group ΝRcRd in which Rc and Rd are as defined above; or a group CONReRf in which Re and Rf are as defined above;
1) a C3-1ocycloalkyl group which may be monocyclic, bicyclic or tricyclic and optionally may be bridged and is optionally substituted by one or more carboxy; fluoro; hydroxy; a
C1-3alkoxy group optionally substituted on C2 or C3 by carboxy; a group NReRf in which5 Re and Rf are as defined above; or a group CONReRf in which Re and Rf are as defined above; m) a C1-6alkoxycarbonyl group;
B) a heteroaryl group which is optionally substituted by groups i) to xxix) as described for phenyl above; Q C) a group of formula NRcRd in which Rc and Rd are as defined above;
D) a C3-7cycloalkyl group optionally substituted by one or more hydroxy or a group of formula NReRf in which Re and Rf are as defined above; E) a carbon linked saturated or partially unsaturated 4 to 8 membered heterocyclic group containing one or more N, S or O, wherein the S may be in its oxidised form of SO or SO2, which is optionally fused to a benz ring and/or is optionally substituted by one or more of the following: hydroxy; oxo; a C1-6alkoxy group; carboxy; hydroxy; C^alkanoyl;
5 a Cμδalkylsulfonyl group; amino; Ci-3alkylamino; di(Ci-3 alkyl)amino; or a C^alkyl optionally substituted by one or more hydroxy or C^alkoxy;
F) a C1.6 alkoxycarbonyl group;
G) a C2-6alkynyl group:
H) a group -C0NR°Rd in which R° and Rd are as defined above; o I) a Cμβalkoxy group;
J) a C2-6alkenyl group:
K) a C^alkyl group;
L) a Ci-βalkylsulphonyl group;
M) phenylsulfonyl; s N) heteroarylsulfonyl;
O) benzoyl;
P) a Ci-βalkanoyl group
Q) hydroxy;
R) oxo; o S) carboxy;
T) fluoro or R1 represents
2) a C3-7cycloalkyl group optionally substituted by one or two groups selected from A to T above; 5 3) a C2-6alkynyl group optionally substituted by one or two groups selected from A to T above;
4) a carbon linked saturated or partially unsaturated 4 to 8 membered heterocyclic group containing one or more N, S or O, wherein the S may be in its oxidised form of SO or
SO2, which is optionally fused to a benz ring and any ring is optionally substituted by0 a group A to T as defined above one or more of the following: hydroxy, oxo, carboxy, a
C^alkoxy group, hydroxy, a d-βalkylsulfonyl group, Ci-4alkanoyl, benzoyl, amino, G1. 3alkylamino, (Ii(C1-3 alkyl)amino or a Ci^alkyl optionally substituted by one or more hydroxy or C1-6alkoxy;
5) a C2-6alkenyl group optionally substituted by one or two groups selected from A to T above; 6) optionally substituted phenyl including optional fusion of the phenyl ring to a saturated or partially unsaturated 5 to 6 membered heterocyclic ring optionally containing one, two or three hetero atoms selected from oxygen, sulphur optionally in its oxidised forms of SO or SO2 or nitrogen wherein the heterocyclic ring is optionally substituted by one or more of the following: a C^alkoxy group; a Ci-6alkanoyl group;carboxy; a C1-6alkylsulfonyl group; a Ci-βalkoxycarbonyl group; carbamoyl; N-Ci-βalkylcarbamoyl; N, N-diQ. βalkylcarbamoyl; hydroxy; oxo; a Ci^alkyl group (which is optionally substituted by one or more of the following: a Q-βalkoxy group, hydroxy or a group of formula ΝRcRd in which R° and Rd are as defined above) and wherein the phenyl ring is optionally substituted by one or more of the groups i to xxix listed above or by a heteroaryl group optionally substituted by one or more groups i) to xxix) above or by an ureido group of formula RmRnN-C(O)-NH- in which Rm and Rn independently represent H, a C1-6alkyl group optionally substituted by a
Figure imgf000006_0001
group, or Rm and Rn together with the nitrogen atom to which they are attached represent a saturated or partially unsaturated 4 to 8 membered heterocyclic ring optionally containing an additional sulphur including oxidised as SO or SO2 , oxygen or nitrogen and/or optionally fused to a benz ring and/or optionally substituted by one or more of the following: a d-βalkoxy group; hydroxy; oxo; carboxy; a Ci-6alkylsulfonyl group; or a C^alkyl group optionally substituted by one or more hydroxy or C^alkoxy; 7) optionally substituted heteroaryl including N-oxides and S-oxides thereof optionally substituted by one or more of the groups i to xxi listed above; wherein any alkyl chain mentioned in any of the definitions from A to P above or in any of the definitions i to xxix above is optionally substituted by 1) one group selected from: carboxy; hydroxy; a C1-3alkoxy group optionally substituted on C2 or C3 by carboxy; a group NRcRd in which Rc and Rd are as defined above; or a group CONReRf in which Re and Rf are as defined above; and /or by 2) from one to five fluoro; and further wherein any cycloalkyl, phenyl, heteroaryl ring or carbon linked saturated or partially saturated 4 to 8 membered heterocyclic group in the list of optional substituents from A to P above or in any of the definitions i to xxix above, for which specific substitution has not been previously mentioned, is optionally substituted by one group selected from: carboxy; hydroxy; a Ci-3alkoxy group optionally substituted on C2 or C3 by carboxy; a group NRcRd in which R° and Rd are as defined above; or a group CONReRf in which Re and Rf are as defined above; and /or is optionally substituted by one to five fluoro;
R2 represents H, halo, a Ci_3alkyl group, trifluoromethoxy or cyano; R3 represents H, halo, a Ci-3alkyl group, trifluoromethoxy or cyano; R4 represents i) H, ii) a C^alkyl group optionally substituted by one or more halo iii) a C1- 3alkoxy group optionally substituted by one or more halo iv) halo, v) nitro, vi) cyano, vii) a C1-6alkylS(O)y(O)z- wherein y is 0,1 or 2 and z is 0 except when y is 2 when z is 0 or 1 viii) a group CH2NRURV in which Ru and Rv independently represent H; a C1-3alkylsulphonyl group, a C1-3alkanoyl group or a Ci_3alkyl group or Ru and Rv together with the nitrogen atom to which they are attached represent azetidinyl, pyrrolidinyl, piperidinyl or morpholinyl; ix) a group CO2RW in which Rw is a Ci-3alkyl group; or x) a group CONRxRy in which Rx and Ry independently represent H; or a C1-3alkyl group or Rx and Ry together with the nitrogen atom to which they are attached represent azetidinyl; pyrrolidinyl, piperidinyl or morpholinyl; R5 and R5 independently represent H, halo, cyano, C1-3alkyl optionally substituted by one or more halo or C1-3alkoxy optionally substituted by one or more halo;
R6 and R6 independently represent H, halo, cyano, C1-3alkyl optionally substituted by one or more halo or C1-3alkoxy optionally substituted by one or more halo; and
R7 is H or OH.
In a first group of compounds of formula I, R7 is H. In a particular group of compounds of formula II
Figure imgf000007_0001
or a pharmaceutically acceptable salt thereof, in which
R1 represents 1) a C^alkyl group optionally substituted by one or more of the following: a) phenyl b) pyridyl or c) a Ci-3alkoxy group 2) a C3-7cycloalkyl group or 3) phenyl ; R2 represents H, halo, a Ci-3alkyl group; or trifluoromethoxy; R3 represents H, halo, a C1-3alkyl group; or trifluoromethoxy; provided that one of R2 and R3 is other than H; R4 represents cyano.
Further sub-definitions of the meaning of R1, R2, R3, R4, R5, R5' R6, R6'and R7 in compounds of formula I now follow. It will be understood that any combination of these sub-definitions may be used instead of the original definitions where appropriate in any of the compound groups, claims or embodiments defined hereinbefore or hereinafter. In one group of compounds of formula I or of formula HA, R1 represents a Ci_6alkyl group optionally substituted by 3-pyridyl or 4-pyridyl. In a second group of compounds of formula I or of formula HA, R2 represents methyl and R3 is H.
"Pharmaceutically acceptable salt", where such salts are possible, includes both pharmaceutically acceptable acid and base addition salts. A suitable pharmaceutically acceptable salt of a compound of formula I is, for example, an acid-addition salt of a compound of formula I which is sufficiently basic, for example an acid-addition salt with an inorganic or organic acid such as hydrochloric, hydrobromic, sulphuric, trifluoroacetic, citric or maleic acid; or, for example a base-addition salt of a compound of formula I which is sufficiently acidic, for example an alkali or alkaline earth metal salt such as a sodium, calcium or magnesium salt, or an ammonium salt, or a salt with an organic base such as methylamine, dimethylamine, trimethylamme, piperidine, morpholine or tris-(2-hydroxyethyl)amine.
Throughout the specification and the appended claims, a given chemical formula or name shall encompass all stereo and optical isomers and racemates thereof as well as mixtures in different proportions of the separate enantiomers, where such isomers and enantiomers exist, as well as pharmaceutically acceptable salts thereof and solvates thereof such as for instance hydrates including solvates of the free compounds or solvates of a salt of the compound. Isomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation. The enantiomers may be isolated by separation of racemate for example by fractional crystallisation, resolution or HPLC. The diastereomers may be isolated by separation of isomer mixtures for instance by fractional crystallisation, HPLC or flash chromatography. Alternatively the stereoisomers may be made by chiral synthesis from chiral starting materials under conditions that will not cause racemisation or epimerisation, or by derivatisation, with a chiral reagent. All stereoisomers are included within the scope of the invention. AU tautomers, where possible, are included within the scope of the invention. The present invention also encompasses compounds containing one or more isotopes for example 14C, 11C or 19F and their use as isotopically labelled compounds for pharmacological and metabolic studies. The present invention also encompasses prodrugs of a compound of formula I that is compounds which are converted into a compound of formula I in vivo. The following definitions shall apply throughout the specification and the appended claims.
The term "C3-iocycloalkyl group which may be monocyclic, bicyclic or tricyclic and optionally may be bridged" includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, bicyclo(2.2.1)heptyl, bicyclo(2.2.2)octyl, perhydroindanyl and adamantyl.
The term "heteroaryl" includes pyrrolyl, thienyl, furyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiadiazolyl, 1,3,4-oxadiazolyl, 1,2,4- oxadiazolyl, triazolyl, furazanyl, tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, 1,3,5-triazinyl quinolyl, isoquinolyl, benzthienyl, benzofuranyl, benzofurazanyl, benzoxazolyl, benzimidazolyl, indolyl, benzthiazolyl, indazolyl, cinnolinyl, quinazolinyl, quinoxalinyl, phthalazinyl, 1,5-naphthyridinyl, 1,6-naphthyridinyl, 1,7-naphthyridinyl, 1,8- naphthyridinyl, pyrrolopyridinyl, pyrrolopyrazinyl, pyrazolopyridinyl or imidazopyridinyl. The term "heteroaryl including N-oxides" includes heteroaryls as described immediately above and in addition N-oxides of such heteroaryls where such N-oxides are known to those skilled in the art to exist and are known to be stable at ambient conditions for example pyridine-N-oxides.
The term "a carbon linked saturated or partially saturated 3 to 8 membered heterocyclic group containing one or more Ν, S, SO, SO2 or O" includes oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, 2,3-dihydro-l,3-thiazolyl, 1,3-thiazolidinyl, 1,3-oxazolidinyl, oxepanyl, azetidinyl, pyrrolinyl, pyrrolidinyl, morpholinyl, thiamorpholinyl (perhydro-l,4-thiazinyl), (8-oxa-3-azabicyclo[3.2.1]octyl), (7-oxa-3- azabicyclo[3.1.1 ]heptyl), perhydroazepinyl, perhydrooxazepinyl, tetrahydro- 1 ,4-thiazinyl, 1-oxotetrahydrothienyl, l,l-dioxotetrahydro-l,4-thiazinyl, piperidinyl, homopiperidinyl, piperazinyl, homopiperazinyl, dihydropyridinyl, tetrahydropyridinyl, dihydropyrimidinyl or tetrahydropyrimidinyl each of which amy be optionally substituted as previously described.
When two substituents on an amine together with the nitrogen atom to which they are attached represent a saturated or partially unsaturated 3 to 8 membered heterocyclic ring optionally containing an additional oxygen, sulphur, SO, SO2 or nitrogen O and/or optionally fused to a benz ring then such rings include azetidino, pyrrolidino, morpholino, piperidino, imidazolidinyl, imidazolinyl, piperazino, thiamorpholino (perhydro-1,4- thiazinyl), homopiperazino, perhydroazepino, perhydrooxazepino, (2,3-dihydro-l,3- thiazolyl, 1,3-thiazolidinyl, 1,3-oxazolidinyl, oxepanyl , oxazepanyl, dihydropyrimidinyl, tetrahydropyrimidinyl, and homopiperidinyl, each of which is optionally substituted as previously described.
Unless otherwise stated or indicated, the term "alkyl" denotes either a straight or branched alkyl group. Examples of said alkyl include methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl, sec-butyl , t-butyl, pentyl, isopentyl, neopentyl, tert-pentyl, hexyl and isohexyl. Preferred alkyl groups are methyl, ethyl, propyl, isopropyl, butyl and tertiary butyl.
Unless otherwise stated or indicated, the term "alkoxy" denotes a group O-alkyl, wherein alkyl is as defined above.
Unless otherwise stated or indicated, the term "halogen" shall mean fluorine, chlorine, bromine or iodine. An
Figure imgf000010_0001
is acetoxy. Examples of "Cμδalkoxycarbonyl" include C]-4alkoxycarbonyl, methoxycarbonyl, ethoxycarbonyl, n- and t-butoxycarbonyl. Examples of "C1-6alkoxycarbonylamino" include methoxycarbonylamino, ethoxycarbonylamino, n- and ^-butoxycarbonylamino. Examples of "C^alkoxy" include methoxy, ethoxy and propoxy. Examples of "Cμβalkanoylamino" include formamido, acetamido and propionylamino. Examples of "C1-6alkylS(O)a wherein a is 0 to 2" include Ci-4alkylsulphonyl, methylthio, ethylthio, methylsulphinyl, ethylsulphinyl, mesyl and ethylsulphonyl. Examples of "C^alkylsulphonylamino" include methylsulphonylamino, ethylsulphonylamino and propylsulphonylamino. Examples of "Ci-6alkylsulphonyl-N-(C1-6alkyl)amino" include methylsulphonyl-N-methylamino, ethylsulphonyl-N-methylamino and propylsulphonyl-N-ethylamino. Examples of "Ci-ealkanoyl" include C1-4alkanoyl, propionyl and acetyl. Examples of
Figure imgf000011_0001
include methylamino and ethylamino. Examples of "N,N-(Ci-6alkyl)2amino" include di-N-methylamino, di-(N-ethyl)amino and N-ethyl-N-methylamino. Examples of "C2-6alkenyl" are vinyl, allyl and 1-propenyl. Examples of "C2-6alkynyl" are ethynyl, 1-propynyl and 2-propynyl. Examples of "N-(Ci-6alkyl)sulphamoyl" are N-(methyl)sulphamoyl and N-(ethyl)sulphamoyl. Examples of "N-(Ci-6alkyl)2Sulphamoyl" are N,N-(dimethyl)sulphamoyl and
N-(methyl)-N-(ethyl)sulphamoyl. Examples of "^(C^ealky^carbamoyl" are N-(Ci-4alkyl)carbamoyl, methylaminocarbonyl and ethylaminocarbonyl. Examples of "N,N-(Ci.6alkyl)2carbamoyl" are N,N-(C]-4alkyl)carbamoyl, dimethylaminocarbonyl and methylethylaminocarbonyl. Examples of "C3-8Cy cloalkyl ring" are cyclopropyl and cyclohexyl. Examples of "(heterocyclic group)C1-6alkyl" include pyridylmethyl,
3-morpholinopropyl and 2-pyrimid-2-ylethyl. Examples of "C3-8cycloalkylCi-6cycloalkyl" include cyclopropylmethyl and 2-cyclohexylpropyl. "N-(Ci-6alkyl)sulphamoylamino" are N-(methyl)sulphamoylamino andN-(ethyl)sulphamoylamino. Examples of "N-(C1-6alkyl)2sulphamoylamino" are N,N-(dimethyl)sulphamoylamino and N-(methyl)-N-(ethyl)sulphamoylamino. Examples of "Ci-ealkylsulphonylaminocarbonyl" include methylsulphonylaminocarbonyl, ethylsulphonylaminocarbonyl and propylsulphonylaminocarbonyl.
Specific compounds of the invention include one or more including any combination of the following compounds below labelled as List 1: 2-methylpropyl Ν-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl- phenyl]carbamate; propan-2-yl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate benzyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate; phenyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate; benzyl N-[2-chloro-5-[4-(4-cyanophenyl)piperidine-l-carbonyl]phenyl]carbamate; benzyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-(trifluoromethoxy)phenyl]- carbamate; benzyl N-[3-[4-(4-cyanophenyl)piperidine-l-carbonyl]-4-methyl-phenyl]carbamate; benzyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-fluoro-phenyl]carbamate; benzyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2,4-dimethyl-phenyl]carbamate;
2-methylpropyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2,4-dimethyl- 5 phenyl] carbamate; pyridin-3-ylmethyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-metliyl- phenyl]carbamate; pyridin-4-ylmethyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl- phenyljcarbamate; i o methyl N- [5 -[4-(4-cyanophenyl)piperidine- 1 -carbonyl] -2-methyl-phenyl] carbamate ;
2-methoxyethyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl- phenyljcarbamate; phenethyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate ethyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate; 15 propyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate; benzyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methoxy-phenyl]carbamate; tert-Butyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate; benzyl N-[5-[4-[4-(dimethylcarbamoyl)phenyl]piperidine-l-carbonyl]-2-methyl- phenyl] carbamate; 20 benzyl N-[5-[4-(4-carbamoylphenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate;
2-methylpropyl N-[2-chloro-5-[4-(4-cyanophenyl)piperidine-l-carbonyl]phenyl]- carbamate; phenyl N-[2-chloro-5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]phenyl]carbamate;
2-methylpropyl N-[3-[4-(4-cyanophenyl)piperidine-l-carbonyl]-4-methylphenyl]- 25 carbamate;
2-methylpropyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-fluorophenyl]carbamate;
2-methylpropyl N-[3-[4-(4-cyanophenyl)piperidine-l-carbonyl]phenyl]carbamate; and benzyl N-[3-[4-(4-cyanophenyl)piρeridine- 1 -carbonyl]phenyl] carbamate or a pharmaceutically-acceptable salt thereof. 30 A compound of the Formula I, or a pharmaceutically-acceptable salt thereof, may be prepared by any process known to be applicable to the preparation of chemically-related compounds. Such processes, when used to prepare a compound of the formula I are provided as a further feature of the invention and are illustrated by the following representative process variants. Necessary starting materials may be obtained by standard procedures of organic chemistry. The preparation of such starting materials is described in conjunction with the following representative process variants and within the accompanying Examples. Alternatively necessary starting materials are obtainable by analogous procedures to those illustrated that are within the ordinary skill of an organic chemist.
According to a further aspect of the present invention provides a process for preparing a compound of formula I or a pharmaceutically acceptable salt thereof (wherein R1, R2, R3, R4, R5, R5', R6, R6, and R7 are, unless otherwise specified, as defined in formula I) which process comprises : (a) reacting a compound of formula VI
Figure imgf000013_0001
with a compound of formula VII R1OC(O)X
VII wherein X is a leaving group, for example halo, e.g. chloro, in the presence of a diluent, for example an organic liquid, e.g. dichloromethane, optionally in presence of a base, for example pyridine, at a temperature in the range of 0-1500C.
Compounds of formula I may also be prepared by reacting a compound of formula VIII
Figure imgf000014_0001
VIlI with a compound of formula IX
Figure imgf000014_0002
IX in which X represents a leaving group for example halo, e.g. chloro, in the presence of a diluent for example a solvent e.g. dichloromethane and optionally in the presence of a base, for example an organic amine e.g DIPEA, at a temperature in the range of 0-1500C. Compounds of formula I may also be prepared by reacting a compound of formula VIII
Figure imgf000014_0003
VIII with a compound of formula X
Figure imgf000014_0004
X in which X represents a replaceable group, eg. Cl, Br, I, OMesyl, or OTriflyl in the presence of carbon monoxide and in the presence of a metal catalyst, eg. Pd or derivatives thereof, and in a solvent such as an alcohol, THF, toluene, or DMF, and in the temperature range 0 - 15O0C. The carbon monoxide may be gaseous or in the form of a metal carbonyl, eg. Molybdenum hexacarbonyl. Compounds of formula I may also be prepared by reacting a compound of formula VIII
Figure imgf000015_0001
VIII with a compound of formula XI
Figure imgf000015_0002
Xl optionally in the presence of a coupling agent and optionally in the presence of a diluent for example a solvent at a temperature in the range of 0-15O0C.
Examples of coupling agents are Dichlorotriphenyl phosphorane (DCTPP), 1-ethyl-
3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDAC), O-benzotriazol-1-yl-
N,N,N',N'-tetramethyluronium hexafluorophosphate (HTBU), O-(7-azabenzotriazol-l-yl)- N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU) and 4-(4,6-dimethoxy-l ,3,5- triazin-2-yl)-4-methylmorpholinium chloride (DMTMM).
Examples of optional additives are: 1 -hydroxy benzotriazole (HOBt), 4- dimethylamino pyridine (DMAP), di-ωo-propylethylamine (DIPEA), and triethylamine
(TEA). Examples of suitable solvents are: dimethyl formamide (DMF), chloroform, dichloromethane (DCM), and tetrahydrofuran (THF). Certain compounds of formula I may be converted into other compounds of formula I by methods known to those skilled in the art. For example, compounds of formula I
Compounds of formula I in which R1 represents an optionally substituted pyridyl- s N-oxide may be prepared by reacting a compound of formula I in which R1 represents an optionally substituted pyridyl with an oxidising agent for example urea hydrogen peroxide or 3-chloroperbenzoic acid, in the presence of a diluent for example dichloromethane or acetonitrile at a temperature in the range of 0-150°C.
In other processes compounds of formula I containing a sulphide group may be I0 oxidised to SO or SO2 for example by use of potassium peroxymonosulfate, nitriles may be reduce to aniinomethyl compounds, amines may be acylated or sulphonated to give amides or sulphonamides, respectively, activated heteroaryl halides may be hydrolysed to hydroxy groups, and esters may be hydrolysed to acids.
It will be appreciated by those skilled in the art that certain functional groups may require is protection before certain transformations are attempted followed by deprotection after the particular transformation. Such methods are well known to those skilled in the art and are described in "Protective Groups in Organic Synthesis", 2 Edition (1991) by Greene and
Wuts.
Certain intermediates of formula VI are believed to be novel and are herein claimed
20 as another aspect of the present invention. Pharmaceutical preparations
The compounds of the invention will normally be administered via the oral, parenteral, intravenous, intramuscular, subcutaneous or in other injectable ways, buccal, rectal, vaginal, transdermal and/or nasal route and/or via inhalation, in the form of
25 pharmaceutical preparations comprising the active ingredient or a pharmaceutically acceptable addition salt, in a pharmaceutically acceptable dosage form. Depending upon the disorder and patient to be treated and the route of administration, the compositions may be administered at varying doses.
Suitable daily doses of the compounds of the invention in the therapeutic treatment
3o of humans are about 0.001-10 mg/kg body weight, preferably 0.01-1 mg/kg body weight. Oral formulations are preferred particularly tablets or capsules which may be formulated by methods known to those skilled in the art to provide doses of the active compound in the range of 0.5mg to 500mg for example 1 mg, 3 mg, 5 mg, 10 mg, 25mg, 50mg, lOOmg and 250mg.
According to a further aspect of the invention there is also provided a pharmaceutical formulation comprising a compound of formula I, or pharmaceutically acceptable salt thereof, including the compound of the proviso, in admixture with pharmaceutically acceptable adjuvants, diluents and/or carriers. Pharmacological properties
The compounds of formula (I) are useful for the treatment of obesity or being overweight, (e.g., promotion of weight loss and maintenance of weight loss), prevention of weight gain (e.g., medication-induced or subsequent to cessation of smoking), for modulation of appetite and/or satiety, eating disorders (e.g. binge eating, bulimia and compulsive eating), dyslipidaemia and the treatment of type 2 diabetes mellitus.
The present compounds of formula (I) are useful for the prophylaxis and/or treatment of clinical conditions associated with inherent or induced reduced sensitivity to insulin (insulin resistance) and associated metabolic disorders (also known as the metabolic syndrome). These clinical conditions will include, but will not be limited to, general obesity, abdominal obesity, arterial hypertension, hyperinsulinaemia, hyperglycaemia, type 2 diabetes and the dyslipidaemia characteristically appearing with insulin resistance. This dyslipidaemia, also known as the atherogenic lipoprotein profile, is characterised by moderately elevated non-esterified fatty acids, elevated very low density lipoprotein (VLDL) triglyceride rich particles, high Apo B levels, low high density lipoprotein (HDL) levels associated with low apoAI particle levels and high Apo B levels in the presence of small, dense, low density lipoproteins (LDL) particles, phenotype B. The compounds of the present invention are expected to be useful in treating patients with combined or mixed hyperlipidemias or various degrees of hypertriglyceridemias and postprandial dyslipidemia with or without other manifestations of the metabolic syndrome.
Treatment with the present compounds is expected to lower the cardiovascular morbidity and mortality associated with atherosclerosis due to their antidyslipidaemic as well as anti-inflammatory properties. The cardiovascular disease conditions include macro- angiopathies of various internal organs causing myocardial infarction, congestive heart failure, cerebrovascular disease and peripheral arterial insufficiency of the lower extremities. Because of their insulin sensitizing effect the compounds of formula I are also expected to prevent or delay the development of type 2 diabetes from the metabolic syndrome and diabetes of pregnancy. Therefore the development of long-term complications associated with chronic hyperglycaemia in diabetes mellitus, such as the micro-angiopathies causing renal disease, retinal damage and peripheral vascular disease of the lower limbs, is expected to be delayed. Furthermore the compounds may be useful in treatment of various conditions outside the cardiovascular system whether or not associated with insulin resistance, like polycystic ovarian syndrome, obesity, cancer and states of inflammatory disease including neurodegenerative disorders such as mild cognitive impairment, Alzheimer's disease, Parkinson's disease and multiple sclerosis.
The compounds of formula I may also be useful in the treatment of metabolic syndrome and Prader-Willi syndrome.
In another aspect the present invention provides a compound of formula I as previously defined for use as a medicament. In a further aspect the present invention provides the use of a compound of formula
I in the preparation of a medicament for the treatment or prophylaxis of obesity or being overweight, (e.g., promotion of weight loss and maintenance of weight loss), prevention of weight gain (e.g., medication-induced or subsequent to cessation of smoking), for modulation of appetite and/or satiety, eating disorders (e.g. binge eating, bulimia and compulsive eating) and for the treatment or prophylaxis of dyslipidaemia and for the treatment or prophylaxis of type 2 diabetes mellitus.
In a still further aspect the present invention provides a method of treating obesity or being overweight, (e.g., promotion of weight loss and maintenance of weight loss), prevention of weight gain (e.g., medication-induced or subsequent to cessation of smoking), for modulation of appetite and/or satiety, eating disorders (e.g. binge eating, bulimia and compulsive eating) dyslipidaemia and type 2 diabetes mellitus comprising administering a pharmacologically effective amount of a compound of formula I, including the compound of the proviso, to a patient in need thereof. Combination Therapy The compounds of the invention may be combined with another therapeutic agent that is useful in the treatment of obesity such as other anti-obesity drugs, that affect energy expenditure, glycolysis, gluconeogenesis, glucogenolysis, lipolysis, lipogenesis, fat absorption, fat storage, fat excretion, hunger and/or satiety and/or craving mechanisms, appetite/motivation, food intake, or G-I motility.
The compounds of the invention may further be combined with another therapeutic agent that is useful in the treatment of disorders associated with obesity such as hypertension, hyperlipidaemias, dyslipidaemias, diabetes, sleep apnea, asthma, heart disorders, atherosclerosis, macro and micro vascular diseases, liver steatosis, cancer, joint disorders, and gallbladder disorders. For example, a compound of the present invention may be used in combination with a another therapeutic agent that lowers blood pressure or that decreases the ratio of LDL:HDL or an agent that causes a decrease in circulating levels of LDL-cholesterol. In patients with diabetes mellitus the compounds of the invention may also be combined with therapeutic agents used to treat complications related to microangiopathies.
The compounds of the invention may be used alongside other therapies for the treatment of obesity and its associated complications the metabolic syndrome and type 2 diabetes, these include biguanide drugs, insulin (synthetic insulin analogues) and oral antihyperglycemics (these are divided into prandial glucose regulators and alpha- glucosidase inhibitors).
In another aspect of the invention, the compound of formula I, or a pharmaceutically acceptable salt thereof may be administered in association with a PPAR modulating agent. PPAR modulating agents include but are not limited to a PPAR alpha and/or gamma agonist, or pharmaceutically acceptable salts, solvates, solvates of such salts or prodrugs thereof. Suitable PPAR alpha and/or gamma agonists, pharmaceutically acceptable salts, solvates, solvates of such salts or prodrugs thereof are well known in the art. In addition the combination of the invention may be used in conjunction with a sulfonylurea. The present invention also includes a compound of the present invention in combination with a cholesterol-lowering agent. The cholesterol-lowering agents referred to in this application include but are not limited to inhibitors of HMG-CoA reductase (3- hydroxy-3-methylglutaryl coenzyme A reductase). Suitably the HMG-CoA reductase inhibitor is a statin.
In the present application, the term "cholesterol-lowering agent" also includes chemical modifications of the HMG-CoA reductase inhibitors, such as esters, prodrugs and metabolites, whether active or inactive.
The present invention also includes a compound of the present invention in combination with an inhibitor of the ileal bile acid transport system (IBAT inhibitor). The present invention also includes a compound of the present invention in combination with a bile acid binding resin.
The present invention also includes a compound of the present invention in combination with a bile acid sequestering agent, for example colestipol or cholestyramine or cholestagel. According to an additional further aspect of the present invention there is provided a combination treatment comprising the administration of an effective amount of a compound of the formula I, or a pharmaceutically acceptable salt thereof, optionally together with a pharmaceutically acceptable diluent or carrier, with the simultaneous, sequential or separate administration one or more of the following agents selected from: a CETP (cholesteryl ester transfer protein) inhibitor; a cholesterol absorption antagonist; a MTP (microsomal transfer protein) inhibitor ; a nicotinic acid derivative, including slow release and combination products; a phytosterol compound ; probucol; an anti-coagulant; an omega-3 fatty acid ; another anti-obesity compound for example sibutramine, phentermine, orlistat, bupropion, ephedrine, thyroxine; an aldose reductase inhibitor; a glycogen phosphorylase inhibitor; a glycogen synthase kinase inhibitors; a glucokinase activator; a haemostasis modulator; an antithrombotic; an activator of fibrinolysis; an antiplatelet agent; a thrombin antagonist; a factor Xa inhibitor; - a factor Vila inhibitor; an antiplatelet agents; a 5 HT transporter inhibitor; an antihypertensive compound for example an angiotensin converting enzyme (ACE) inhibitor, an angiotensin II receptor antagonist, an adrenergic blocker, an alpha adrenergic blocker, a beta adrenergic blocker, a mixed alpha/beta adrenergic blocker, an adrenergic stimulant, calcium channel blocker, an AT-I blocker, a saluretic, a diuretic or a vasodilator; a melanin concentrating hormone (MCH) modulator; an NPY receptor modulator; for example an NPY agonist or an NPY2 agonist or an NPY5 antagonist; an Mc4r modulator for example an Mc4r agonist; an Mc3r modulator for example an Mc3r agonist; an orexin receptor modulator for example an antagonist; a phosphoinositide-dependent protein kinase (PDK) modulator; or modulators of nuclear receptors for example LXR, FXR, RXR, GR, ERRα, β, PPARα, β, γ, δ and RORalpha; a monoamine transmission-modulating agent, for example a selective serotonin reuptake inhibitor (SSRI), a noradrenaline reuptake inhibitor (NARI), a noradrenaline-serotonin reuptake inhibitor (SNRI), a monoamine oxidase inhibitor (MAOI), a tricyclic antidepressive agent (TCA), a noradrenergic and specific serotonergic antidepressant
(NaSSA); an antipsychotic agent for example olanzapine and clozapine; a serotonin receptor modulator; a leptin/leptin receptor modulator; a CBl receptor modulator for example an inverse agonist or an antagonist; a GLK receptor modulator; a DPP-IV inhibitor; a cholesterol absorption inhibitor; a GLP-I agonist; an SGLT-2 inhibitor; a DGATl inhibitor; a DGAT2 inhibitor; a DGAT2 inhibitor anti-sense oligonucleotide; a ghrelin antibody; a ghrelin antagonist; an l lβ HSD-I inhibitor; an UCP- 1,2 or 3 activator; or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, optionally together with a pharmaceutically acceptable diluent or carrier to a warmblooded animal, such as man in need of such therapeutic treatment. According to an additional further aspect of the present invention there is provided a combination treatment comprising the administration of an effective amount of a compound of the formula I, or a pharmaceutically acceptable salt thereof, optionally together with a pharmaceutically acceptable diluent or carrier, with the simultaneous, sequential or separate administration of very low calorie diets (VLCD) or low-calorie diets (LCD).
Therefore in an additional feature of the invention, there is provided a method for the treatment of obesity and its associated complications in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof in simultaneous, sequential or separate administration with an effective amount of a compound from one of the other classes of compounds described in this combination section, or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof.
Therefore in an additional feature of the invention, there is provided a method of treating hyperlipidemic conditions in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof in simultaneous, sequential or separate administration with an effective amount of a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof. According to a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, in association with a pharmaceutically acceptable diluent or carrier.
According to a further aspect of the present invention there is provided a kit comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof.
According to a further aspect of the present invention there is provided a kit comprising: a) a compound of formula I, or a pharmaceutically acceptable salt thereof, in a first unit dosage form; b) a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof; in a second unit dosage form; and c) container means for containing said first and second dosage forms.
According to a further aspect of the present invention there is provided a kit comprising: a) a compound of formula I, or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable diluent or carrier, in a first unit dosage form; b) a compound from one of the other classes of compounds described in this combination section or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, in a second unit dosage form; and c) container means for containing said first and second dosage forms.
According to another feature of the invention there is provided the use of a compound of the formula I, or a pharmaceutically acceptable salt thereof, and one of the other compounds described in this combination section, or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, in the manufacture of a medicament for use in the the treatment of obesity and its associated complications in a warm-blooded animal, such as man. According to another feature of the invention there is provided the use of a compound of the formula I, or a pharmaceutically acceptable salt thereof, and one of the other compounds described in this combination section, or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, in the manufacture of a medicament for use in the treatment of hyperlipidaemic conditions in a warm-blooded animal, such as man.
According to a further aspect of the present invention there is provided a combination treatment comprising the administration of an effective amount of a compound of the formula I, or a pharmaceutically acceptable salt thereof, optionally together with a pharmaceutically acceptable diluent or carrier, with the simultaneous, sequential or separate administration of an effective amount of one of the other compounds described in this combination section, or a pharmaceutically acceptable salt, solvate, solvate of such a salt or a prodrug thereof, optionally together with a pharmaceutically acceptable diluent or carrier to a warm-blooded animal, such as man in need of such therapeutic treatment.
Furthermore, a compound of the invention may also be combined with therapeutic agents that are useful in the treatment of disorders or conditions associated with obesity (such as type II diabetes, metabolic syndrome, dyslipidemia, impaired glucose tolerance, hypertension, coronary heart disease, non-alcoholic steatohepatitis, osteoarthritis and some cancers) and psychiatric and neurological conditions.
It will be understood that there are medically accepted definitions of obesity and being overweight. A patient may be identified by, for example, measuring body mass index (BMI), which is calculated by dividing weight in kilograms by height in metres squared, and comparing the result with the definitions. The compounds of the invention may also be useful as anti-cell-proliferation (such as anti-cancer) agents and are therefore useful in methods of treatment of the human or animal body.
Such properties are expected to be of value in the treatment of disease states associated with cell cycle and cell proliferation such as cancers (solid tumors and leukemias), fibroproliferative and differentiative disorders, psoriasis, rheumatoid arthritis, Kaposi's sarcoma, haemangioma, acute and chronic nephropathies, atheroma, atherosclerosis, arterial restenosis, autoimmune diseases, acute and chronic inflammation, bone diseases and ocular diseases with retinal vessel proliferation.
The anti-cancer treatment defined herein may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional surgery or radiotherapy or chemotherapy. Such chemotherapy may include one or more of the following categories of anti-tumour agents:
(i) antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis-platin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan and nitrosoureas); antimetabolites (for example antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and taxotere); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and camptothecin);
(ii) cytostatic agents such as antioestrogens (for example tamoxifen, toremifene, raloxifene, droloxifene and iodoxyfene), oestrogen receptor down regulators (for example fulvestrant), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5α-reductase such as finasteride; (iii) agents which inhibit cancer cell invasion (for example metalloproteinase inhibitors like marimastat and inhibitors of urokinase plasminogen activator receptor function); (iv) inhibitors of growth factor function, for example such inhibitors include growth factor antibodies, growth factor receptor antibodies (for example the anti-erbb2 antibody trastuzumab [Herceptin™] and the anti-erbbl antibody cetuximab [C225]) , farnesyl transferase inhibitors, tyrosine kinase inhibitors and serine/threonine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3- morpholinopropoxy)quinazolin~4-amine, N-(3-ethynylphenyl)-6,7-bis(2- methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774) and 6-acrylamido-N-(3-chloro-4- fluorophenyl)-7-(3-morpholinopropoxy)quinazolin-4-amine (CI 1033), for example inhibitors of the platelet-derived growth factor family and for example inhibitors of the hepatocyte growth factor family;
(v) antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, (for example the anti-vascular endothelial cell growth factor antibody bevacizumab [Avastin™], compounds such as those disclosed in International Patent Applications WO 97/22596, WO 97/30035, WO 97/32856 and WO 98/13354) and compounds that work by other mechanisms (for example linomide, inhibitors of integrin αvβ3 function and angiostatin);
(vi) vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529, WO 00/41669, WO 01/92224, WO 02/04434 and WO 02/08213; (vii) antisense therapies, for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
(viii) gene therapy approaches, including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCAl or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy; and (ix) immunotherapy approaches, including for example ex-vivo and in- vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies.
Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment. Such combination products employ the compounds of this invention within the dosage range described hereinbefore and the other pharmaceutically-active agent within its approved dosage range. The compounds of the present invention may also be useful as anti-infective agents or as anti-bacterial agents.
The compounds of the present invention may also be useful as in decreasing sebum production following topical application. Pharmacological Activity
The compounds of the present invention are Fatty Acid Synthase inhibitors. The activity of the compounds of the invention was demonstrated using the following assay. Human and Rat FAS Enzyme Assay.
Fatty acid synthase is an enzyme complex that harbours seven enzymatic activities catalysing the reductive synthesis of long chain fatty acids from acetyl CoA and malonyl CoA to palmitate. When acetyl CoA and malonyl CoA are forming palmitate NADPH is consumed forming NADP. Since NADPH is fluorescent but not NADP the reaction can be measured by analysing the decrease in fluorescence.
Compounds were added to a black 384 well plate (Matrix) in a volume of 5μl consisting of 20% DMSO and 80% Tris buffer pH 7.5, at a top concentration of ImM. NADPH, 30μl of 166.6μM, formulated in assay buffer (0.1M Tris ph7.5, O.lmM EDTA5ImM glutathione, 0.05%BSA), was then added to all of the wells of the plate. Fatty acid synthase Human or rat enzyme (0.4μg, produced in house), dissolved in 2OmM Tris/HCl pH 7.5, 5mM BOG5ImM TCEP,10% glycerol,lmM EDTA,150mM NaCl, was then added to the plate in a volume of lOμl. Enzyme was added to all but the last two columns of the plate, to which, lOμl of assay buffer was added (0.1M Tris ph7.5, O.lmM EDTA, ImM glutathione, 0.05%BSA) to provide a no enzyme assay control. Following a 15-minute incubation period, at room temperature, the plates were read on an Envision plate reader using 340nm excitation and 460nm emission filters. This served as a time zero background read. Substrates (an equal mix of both malonyl and acetyl CoA) were then added to the plates in a total volume of 5μl. The concentrations of malonyl and acetyl CoA in the mixture were 500μM and 150μM respectively. Both were prepared as 1OmM stock solutions in distilled water and were subsequently diluted to working concentrations in assay buffer. Plates were then incubated for a further 60 minutes, at room temperature, before being read again on the Envision reader using the same parameters as previously used. The data was analysed by subtracting the background time zero data from that generated following the final 60 minute incubate and the percent inhibition compared to the maximum and minimum assay controls was determined. Sigmoid curves were fitted using Origin 7.5 Client software and IC50 values were determined.
The compounds of the present invention were found to inhibit the activation of Fatty Acid Synthase with IC50S in a range of about 0.00 lμM to about 30μM in the above assay. In a preferred range, the examples of the present invention inhibited the activition of Fatty Acid Synthase with IC5oS in a range of about 0.00 lμM to about 5.0μM. In a more preferred range, the compounds inhibit the activation of Fatty Acid Synthase with IC50S in a range of about 0.00 lμM to about O.lμM.
The compound of Example 1 had an ICs0Of 0.4 μM, the compound of Example 3 had an IC50 of 0.4 μM and the compound of Example 7 had an IC50 of 1.7 μM.
The results obtained are given in Table 1 in which Ex. No. stands for Example Number and Inhib (%) stands for the % inhibition at a concentration of lOOμmolar.
Figure imgf000028_0001
The following compounds do not have IC50S in the range of about 0.00 lμM to about 30μM in the above assay: tert-butyl N-[3-[4-(4-cyanophenyl)piperidine-l-carbonyl]phenyl]carbamate 2-methylpropyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2- (trifluoromethoxy)phenyl]carbamate 2-methylpropyl N-[2-methyl-5-[4-(4-methylsulfonylphenyl)piperidine- 1 - carbonyl]phenyl]carbamate and
2-methylpropyl N-[3-[4-(4-cyanophenyl)piperidine-l-carbonyl]-4-fluorophenyl]carbamate These compounds are excluded from the claims of the present application by means 5 of a proviso.
The invention will now be illustrated by the following non-limiting examples in which, unless stated otherwise:
(i) temperatures are given in degrees Celsius (0C); operations were carried out at room or ambient temperature, that is, at a temperature in the range of 18-25 0C, unless otherwiseo stated;
(ii) organic solutions were dried over anhydrous magnesium sulfate; evaporation of solvent was carried out using a rotary evaporator under reduced pressure (600-4000 Pascals;
4.5-30 mmHg) with a bath temperature of up to 60 °C;
(iii) chromatography means flash chromatography on silica gel; thin layer chromatographys (TLC) was carried out on silica gel plates;
(iv) in general, the course of reactions was followed by TLC and / or analytical LC-MS, and reaction times are given for illustration only;
(v) final products had satisfactory proton nuclear magnetic resonance (NMR) spectra and/or mass spectral data; o (vi) yields are given for illustration only and are not necessarily those which can be obtained by diligent process development; preparations were repeated if more material was required;
(vii) when given, NMR data is in the form of delta values for major diagnostic protons, given in parts per million (ppm) relative to tetramethylsilane (TMS) as an internal standard5 when the solvent is CDCl3 (when the solvent is d6--DMSO, it locks on to the 2.49 DMSO peak), determined at 300 MHz unless otherwise indicated; the following abbreviations have been used: s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; b, broad;
(viii) chemical symbols have their usual meanings; SI units and symbols are used;
(ix) solvent ratios are given in volume:volume (v/v) terms; and 0 (x) mass spectra (MS) were run with an electron energy of 70 electron volts in the chemical ionization (CI) mode using a direct exposure probe; where indicated ionization was effected by electron impact (EI), fast atom bombardment (FAB) or electrospray (ESP); values for m/z are given; generally, only ions which indicate the parent mass are reported; and unless otherwise stated, the mass ion quoted is MH+;
[A] When Cl is present in the molecule, the m/z value for the (M+H)+ molecular ion is based on the 35Cl isotope. When there are multiple chlorine atoms in the molecule, the m/z is based on the first peak of the isotope pattern.
(xi) unless stated otherwise compounds containing an asymmetrically substituted carbon and/or sulphur atom have not been resolved;
(xii) where a synthesis is described as being analogous to that described in a previous example the amounts used are the millimolar ratio equivalents to those used in the previous example;
(xvi) the following abbreviations have been used:
ACN Acetonitrile
DIPEA Di-/sø-propylethylamine
EDAC N-ethyl-N'-(3-dimethylaminopropyl)-carbodiimide hydrochloride EtOAc Ethyl acetate
DMA Dimethyl acetamide
DMAP 4-dimethylamino pyridine
DMTMM 4-(4,6-Dimethoxy-l ,3,5-Triazin-2-Yl)-4-Methylmorpholinium
Chloride HATU 0-(7-Azabenzotriazol-l-Yl)-N,N,N',N'-Tetramethyluronium
Hexafluoro-phosphate
HTBU O-benzotriazol- 1 -yl-N,N,N',N'-tetramethyluronium hexafluorophosphate
HOBT 1-Hydroxybenzotriazole NMP N-methyl pyrrolidone
EtOH Ethanol
MeOH Methanol
TEA Triethylamine
TFAA Trifluoroacetic Anhydride THF Tetrahydrofuran Example 1
2-methylpropyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl- phenyljcarbamate
Figure imgf000031_0001
/so-butyl chloroformate (82 μL, 0.63mmol) was added to a stirred suspension of 4-[l-(3- amino-4-methyl-benzoyl)-4-piperidyl]benzonitrile (Intermediate A, 200 mg, 0.63mmol) and pyridine (51 μL, 0.63 mmol) in DCM (5 mL), and the reaction mixture was stirred at ambient temperature for 24hrs. The reaction mixture was then washed sequentially with saturated sodium bicarbonate solution (20 mL), water (20 mL) and brine (20 mL), filtered and reduced in vacuo to give a brown oil which was purified by chromatography (12 g silica column, Optix, eluting with a gradient consisting of 30-70% EtOAc in zsohexane) to give the title compound as a colourless solid (164 mg), 1H NMR (300.072 MHz, CDCl3) δ 0.97 (6H, d),1.57 - 1.92 (4H, m),1.98 (IH, 9),2.28 (3H, s),2.77 - 2.92 (2H, m),2.98 - 3.23 (IH, m),3.95 (2H, d),4.00 - 4.09 (IH, m),4.76 - 4.99 (IH, m),6.53 (IH, s),7.10 - 7.24 (2H, m),7.33 (2H, d),7.60 (2H, d),7.91 (IH, s), m/z 420 (M+H)+. Method 2 Example 2 Propan-2-yl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate
Figure imgf000031_0002
Triphosgene (62 mg, 0.21 mmol) was added to a solution of 4-[l-(3-amino-4-methyl- benzoyl)-4-piperidyl]benzonitrile (Intermediate A, 167 mg, 0.52 mmol ) and DIPEA (180 μL) in anhydrous THF (8 mL). The reaction mixture was purged with nitrogen and stirred at ambient temperature for 3 1A hrs. The reaction mixture was filtered and the filtrate added to a reaction tubes containing 2-propanol (1 mL, large excess). The reaction mixture was then heated with stirring at 65°C for 3hrs, and then concentrated in vacuo. The residue was purified by chromatography (Optix 10™ , 12 g silica column, gradient eluting with hexane containing 40-80% EtOAc) to give the title compound (133 mg, 63%), 1H NMR (300.073 MHz, dmso) δ 1.24 (d, 6H), 1.48 - 1.94 (m, 4H), 2.22 (s, 3H), 2.74 - 3.21 (m, 3H), 3.56 4.00 (m, IH), 4.37 - 4.75 (m, IH), 4.76 - 4.94 (m, IH), 7.06 - 7.14 (m, IH), 7.23 (d, IH), 7.44 (s, 2H), 7.50 (d, 3H), 7.76 (d, 2H), 8.82 (s, IH), m/z 406 (M+H)+.
It will be appreciated that alternative solvents, reagents, additives and conditions may be used in the above reactions. Examples of suitable solvents are THF, DCM, other; examples of bases are TEA, DIPEA and pyridine, and the reactions may be performed at temperatures between O0C and the boiling point of the solvent.
The following compounds were prepared in a manner essentially similar to that described for Example 1, starting from the appropriate intermediate and chloroformate Example 3 benzyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate
Figure imgf000032_0001
Prepared from Intermediate A 1H NMR (300.072 MHz, CDCl3) δ 1.57 - 1.98 (4H, m), 2.27 (3H, s),2.77 - 2.92 (2H, m),2.98 - 3.23 (IH, m), 3.90 - 4.09 (IH, m),4.76 - 4.99 (IH, m), 5.21 (2H, s),6.64 (IH, s),7.10 - 7.21 (2H, m),7.31-7.43 (7H, m),7.61 (2H, d),7.91 (IH, s), m/z 454 (M+H)+. Example 4 phenyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate
Figure imgf000032_0002
Prepared from Intermediate A 1H NMR (300.072 MHz, CDCl3) δ 1.53 - 1.98 (4H, m),2.35 (3H, s),2.75 - 2.89 (2H, m),2.97 - 3.22 (IH, m),3.85 - 4.07 (IH, m),4.71 - 4.97 (IH, m),7.03 (IH, s),7.14 - 7.31 (7H, m),7.36 - 7.43 (2H, m),7.58 (2H, d),7.90 (IH, s), m/z 440 (M+H)+. Example 5 benzyl N-[2-chloro-5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]phenyl]carbamate
Figure imgf000032_0003
Prepared from Intermediate B 1H NMR (300.073 MHz, dmso) δ 1.37 - 2.08 (m, 4H), 2.66 - 3.18 (m, 3H), 3.52 - 3.79
(m, IH), 4.42 - 4.78 (m, IH), 5.16 (s, 2H), 7.21 - 7.28 (m, IH), 7.29 - 7.45 (m, 5H),
7.46 - 7.57 (m, 3H), 7.67 (s, IH), 7.77 (d, 2H), 9.32 (s, IH), m/z 474 (M+H)+ [A].
Example 6 benzyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-
(trifluoromethoxy)phenyl]carbamate
Figure imgf000033_0001
Prepared from Intermediate C 1H NMR (300.072 MHz, CDCl3) δ 1.63 - 1.96 (4H, m),2.79 - 2.94 (2H, m), 3.05 - 3.27 (IH, m), 3.84 - 4.02 (IH, m), 4.77 - 4.97 (IH, m), 5.22 (2H, s),7.10 (IH, s),7.14 - 7.19 (IH, m),7.27 - 7.43 (8H, m),7.62 (2H, d),8.34 (IH, s), m/z 524 (M+H)+. Example 7 benzyl N-[3-[4-(4-cyanophenyl)piperidine-l-carbonyl]-4-methyl-phenyl]carbamate
Figure imgf000033_0002
Prepared from Intermediate D
1H NMR (300.072 MHz, CDCl3) δ 1.46 - 2.00 (4H, m),2.27 (3H, d),2.81 (2H, t),3.07 (IH, t),3.63 (IH, d),4.91 - 4.99 (IH, m),5.18 (2H, s),7.00 - 7.17 (2H, m),7.24 - 7.40 (9H, m),7.60 (2H, d), m/z 454 (M+H)+. Example 8 benzyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-fluoro-phenyl]carbamate
Figure imgf000033_0003
Prepared from Intermediate E 1H NMR (400.132 MHz, DMSO) δ 1.54 - 1.96 (m, 4H), 2.74 - 3.23 (m, 3H), 3.60 - 3.81 (m, IH), 4.50 - 4.73 (m, IH), 5.17 (s, 2H), 7,19 - 7.25 (m, IH), 7.27 - 7.46 (m, 6H), 7.52 (d, 2H), 7.74 - 7.82 (m, 3H), 9.67 (s, IH), m/z 458 (M+H)+. Example 9 benzyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2,4-dimethyl-phenyl]carbamate
Figure imgf000034_0001
Prepared from Intermediate F
1H NMR (300.072 MHz, CDCl3) δl.75 (2H, m), 2.00 (IH, m), 2.22 (3H, s), 2.27 (3H, s), 2.80 - 2.84 (2H, m), 3.10 (IH, m), 3.71 (IH, m), 4.97 (IH, d), 5.20 (2H, s), 6.44 (IH, s), 7.03 (IH, s), 7.34 - 7.40 (7H, m), 7.60 - 7.63 (2H, d), m/z 468 (MH-H)+. Example 10
2-methylpropyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2,4-dimethyl- phenyl] carbamate
Figure imgf000034_0002
Prepared from Intermediate F
1H NMR (300.072 MHz, CDCl3) δθ.95 - 0.98 (6H, d), 1.75 (2H, m), 1.91 - 2.00 (IH, m), 2.24 - 2.25 (6H, s), 2.82 (2H, m), 3.10 (IH, s), 3.72 (IH, m), 3.93 - 3.95 (2H, d), 4.96 (IH, m), 6.34 (IH, s), 7.02 (IH, s), 7.32 (2H, d), 7.61 (2H, d), m/z 434 (M+H)+. Example 11
Pyridin-3 -ylmethyl N- [5 - [4-(4-cyanophenyl)piperidine- 1 -carbonyl] -2-methyl- phenyl] carbamate
Figure imgf000034_0003
Method 2 from Intermediate A
1H NMR (300.073 MHz, dmso) δ 1.46 - 1.96 (m, 4H), 2.22 (s, 3H), 2.77 - 3.18 (m, 3H), 3.56 - 3.98 (m, IH), 4.32 - 4.82 (m, IH), 5.19 (s, 2H), 7.12 (d, IH), 7.25 (d, IH), 7.39 - 7.47 (m, 2H), 7.50 (d, 2H), 7.76 (d, 2H), 7.86 (d, IH), 8.55 (d, IH), 8.65 (s, IH), 9.11 (s, lH), m/z 455 (M+H)+. Example 12
Pyridin-4-ylmethyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl- phenyl]carbamate
Figure imgf000035_0001
Method 2 from Intermediate A 1H NMR (300.073 MHz, dmso) δ 1.48 - 1.93 (m, 4H), 2.25 (s, 3H), 2.79 - 3.17 (m, 3H), 3.58 - 3.96 (m, IH), 4.36 - 4.78 (m, IH), 5.19 (s, 2H), 7.13 (d, IH), 7.26 (d, IH), 7.38 (d, 2H), 7.44 - 7.53 (m, 3H), 7.76 (d, 2H), 8.57 (d, 2H), 9.21 (s, IH), m/z 455 (M+H)+. Example 13 Methyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate
Figure imgf000035_0002
Method 2 from Intermediate A
1H NMR (300.073 MHz, dmso) δ 1.49 - 1.95 (m, 4H), 2.22 (s, 3H), 2.74 - 3.23 (m, 3H), 3.57 - 3.98 (m, 4H), 4.33 - 4.85 (m, IH), 7.11 (d, IH), 7.24 (d, IH), 7.45 (s, IH), 7.50 (d, 2H), 7.76 (d, 2H), 8.93 (s, IH), m/z 378 (M+H)+. Example 14
2-methoxyethyl N-[5-[4-(4-cyanophenyl)piperidme-l-carbonyl]-2-methyl- phenyl]carbamate
Figure imgf000035_0003
Method 2 from Intermediate A 1H NMR (300.073 MHz, dmso) δ 1.49 - 1.92 (m, 4H), 2.23 (s, 3H), 2.76 - 3.22 (m, 3H),
3.29 (s, 3H), 3.56 (t, 2H), 3.63 - 3.97 (m, IH), 4.18 (t, 2H), 4.34 - 4.86 (m, IH), 7.11 (d,
IH), 7.24 (d, IH), 7.43 (s, IH), 7.50 (d, 2H), 7.76 (d, 2H), 9.02 (s, IH), m/z 422
(M+H)+.
Example 15 Phenethyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate
Figure imgf000035_0004
Method 2 from Intermediate A 1H NMR (300.073 MHz, dmso) δ 1.47 - 1.93 (m, 4H), 2.20 (s, 3H), 2.75 - 3.19 (m, 5H),
3.50 - 4.00 (m, IH), 4.27 (t, 2H), 4.38 - 4.83 (m, IH), 7.11 (d, IH), 7.16 - 7.34 (m, 5H),
7.39 (s, IH), 7.50 (d, 2H), 7.76 (d, 2H), 8.92 (s, IH), m/z 468 (M+H)+.
Example 16
Ethyl N-[5-[4~(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate
Figure imgf000036_0001
Method 2 from Intermediate A
1H NMR (300.073 MHz, dmso) δ 1.23 (t, 3H), 1.48 - 1.94 (m, 4H), 2.23 (s, 3H), 2.73 3.22 (m, 3H), 3.56 - 3.98 (m, IH), 4.10 (q, 2H), 4.35 - 4.82 (m, IH), 7.06 - 7.14 (m, IH), 7.24 (d, IH), 7.45 (s, IH), 7.50 (d, 2H), 7.76 (d, 2H), 8.89 (s, IH), m/z 392 (M+H)+. Example 17 Propyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate
Figure imgf000036_0002
Method 2 from Intermediate A
1H NMR (300.073 MHz, dmso) δ 0.92 (t, 3H), 1.49 - 1.94 (m, 6H), 2.23 (s, 3H), 2.76 - 3.22 (m, 3H), 3.55 - 3.94 (m, IH), 4.02 (t, 2H), 4.34 - 4.80 (m, IH), 7.10 (d, IH), 7.24 (d, IH), 7.44 (s, IH), 7.50 (d, 2H), 7.76 (d, 2H), 8.89 (s, IH), m/z 406 (M+H)+. Example 18 Benzyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methoxy-phenyl]carbamate
Figure imgf000036_0003
Method 1 from Intermediate G 1H NMR (300.073 MHz, dmso, 3O0C) δl.51 - 1.70 (2H, m), 1.70 - 1.88 (2H, m), 2.78 -
3.21 (3H, m), 3.83 (3H, s), 3.60 - 4.73 (2H, m), 5.14 (2H, s), 7.06 (IH, d J = 7.6 Hz), 7.14 - 7.20 (IH, m), 7.27 - 7.45 (5H, m), 7.49 (2H, d J = 11.7 Hz), 7.72 - 7.82 (3H, m), 8.70 (IH, s), m/z 470 (M+H)+. Example 19 tert-Butyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate
Figure imgf000037_0001
The title compound was prepared using the method described in Synthetic
Communications 31(21) p3273 (2001), starting from 4- [1 -(3 -amino-4-methyl-benzoyl)-4- piperidyl]benzonitrile (Intermediate A):
1H NMR (300.073 MHz, dmso) δ 1.45 (s, 9H),1.53 - 1.70 (m, 2H), 1.70 - 1.91 (m, 2H),
2.21 (s, 3H),2.68 - 3.00 (m, 1H),3.OO - 3.23 (m, 1H),3.61 - 3.94 (m, 1H),4.21 - 4.83 (m,
1H),7.O4 - 7.12 (m, 1H),7.17 - 7.25 (m, 1H),7.38 (s, 1H),7.5O (d, 2H),7.75 (d, 2H),8.59 (s,
IH) (NB signal due to piperidyl 4-H v. broad and obscured by surrounding signals), m/z
420 (M+H)+, 464 (M+MeCN+H)+.
Example 20
Benzyl N-[5-[4-[4-(dimethylcarbamoyl)phenyl]piperidine-l-carbonyl]-2-methyl- phenyl]carbamate
Figure imgf000037_0002
Method 1 from Intermediate H 1H NMR (300.073 MHz, dmso) δ 1.48 - 1.89 (m, 4H), 2.23 (s, 3H), 2.67 - 3.05 (m, 9H), 3.60 - 3.86 (m, IH), 4.49 - 4.71 (m, IH), 5.14 (s, 2H), 7.12 (d, J= 7.6 Hz, IH), 7.25 (d, J = 7.9 Hz, IH), 7.31 - 7.48 (m, 10H), 9.06 (s, IH), m/z 500 (M+H)+. Example 21 Benzyl N-[5-[4-(4-carbamoylphenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate
Figure imgf000037_0003
Method 1 from Intermediate I 1H NMR (300.073 MHz, dmso) δ 1.49 - 1.91 (m, 4H), 2.23 (s, 3H), 2.69 - 3.17 (m, 3H),
3.50 - 3.99 (m, IH), 4.44 - 4.78 (m, IH), 5.14 (s, 2H), 7.09 - 7.14 (m, IH), 7.22 - 7.27
(m, 2H), 7.31 - 7.44 (m, 7H), 7.46 (s, IH), 7.80 (d, J= 8.1 Hz, 2H), 7.87 (s, IH), 9.06 (s,
IH), m/z 472 (M+H)+. The following Examples were prepared in a similar manner.
Example 22
2-methylpropyl N-[2-chloro-5-[4-(4-cyanophenyl)piperidine-l-carbonyl]phenyl]carbamate
Example 23
Phenyl N-[2-chloro-5-[4-(4-cyanophenyl)piperidine-l-carbonyl]phenyl]carbamate o Example 24
2-methylpropyl N-[3-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-4- methylphenyl] carbamate
Example 25
2-methylpropyl N-[5 -[4-(4-cyanophenyl)piperidine- 1 -carbonyl] -2-fluorophenyl] carbamates Example 26
2-methylpropyl N- [3 - [4-(4-cyanophenyl)piperidine- 1 -carbonyl]phenyl] carbamate
Example 27
Benzyl N-[3-[4-(4-cyanophenyl)piperidine-l-carbonyl]phenyl]carbamate
Preparation of Intermediates o Intermediate A
4-[l-(3-amino-4-methyl-benzoyl)-4-piperidyl]benzonitrile
Figure imgf000038_0001
A solution of 3-amino-4-methyl benzoic acid (4.05 g, 26.792 mmol), 4-(4'-cyanophenyl) piperidine (5 g, 26.79 mmol), N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide 5 hydrochloride [EDAC] (5.64 g, 29.47 mmol, 1.1 eq) and DMAP (328 mg, 2.68 mmol, 0.1 eq) in DMF (60 mL) was stirred at ambient temperature for 2 hrs. Ethyl acetate (200 mL) was added and the resulting solution was washed sequentially with KHSO4 solution (100 mL of 2M), and brine (100ml); a precipitate formed and was filtered off to give the title compound as a colourless solid (5.25 g), 1H NMR (300.073 MHz, DMSO- d6, 30 0C) δ0 1.47 - 1.67 (2H, m), 1.68 - 1.89 (2H, m), 2.05 (3H, s), 2.68 - 3.15 (3H, m), 3.59 - 4.13 (IH, m), 4.22 - 4.76 (IH, m), 4.97 (2H, s), 6.45 - 6.53 (IH, m), 6.63 (IH, s), 6.90 - 6.99 (IH, m)5 7.44 . 7.54 (2H, m), 7.71 - 7.81 (2H, m), m/z 320 (MH-H)+. Intermediate B 4-[l-(3-amino-4-chloro-benzoyl)-4-piperidyl]benzonitrile
Figure imgf000039_0001
The title compound was prepared in a manner similar to that described for Intermediate A, starting from 3-amino-4-chloro-benzoic acid and 4-(4'-cyanophenyl)piperidine; 1H NMR (300.073 MHz, dmso) δ 1.38 - 2.05 (m, 4H), 2.68 - 3.20 (m, 3H), 3.55 - 3.94 (m, IH), 4.30 - 4.79 (m, IH), 5.50 (s, 2H), 6.56 (d, IH), 6.81 (s, IH), 7.22 (d, IH), 7.49 (d, 2H), 7.76 (d, 2H)m/z 340 (M+H)+ [A]. Intermediate C 4-[l-[3-amino-4-(trifluoromethoxy)benzoyl]-4-piperidyl]benzonitrile
Figure imgf000039_0002
3 -nitro-4-(trifluoromethoxy)benzoic acid
Figure imgf000039_0003
4-(trifluoromethoxy) benzoic acid (4 g, 26.3 mmol) was added slowly to a stirred mixture of concentrated sulfuric acid ((6.6 mL, 65.7 mmol) and concentrated nitric acid (4 mL, 44.7 mmol) at 4O0C. When the addition was complete, the reaction mixture was heated to 5O0C and became a pale yellow slurry. After Ih the reaction appeared to have gone to completion and so was poured onto ice and water. A white precipitate formed which was isolated by filtration and washed with water to give the title compound as a colourless crystalline solid (4.2 g), 1H NMR (300.073 MHz, DMSO-d6) δ7.83 - 7.87 (IH, m), 8.32 - 8.36 (IH, m), 8.56 (IH, d), m/z 198 (M+H)+. Step 2; 3-amino-4-(trifluoromethoxy)benzoic acid
Figure imgf000040_0001
A solution of 3-nitro-4-(trifluoromethoxy)benzoic acid (Step 1) (3.51 g, 14 mmol) in MeOH (150 mL) was hydrogenated at ambient temperature and pressure in the presence of 10% palladium on charcoal catalyst (500 mg). The catalyst was removed by filtration and washed through with more MeOH. The filtrate and washings were combined and evaporated to give the title compound as a pale cream solid (2.9 g), 1H NMR (300.073 MHz, dmso) δ 5.60 (br s, 2H),7.07 - 7.23 (m, 2H),7.39 - 7.46 (m, 1H),12.O2 - 13.40 (br s, IH), m/z 220 (M-H)-. Step 3: 4- [ 1 - [3 -amino-4-(trifluoromethoxy)benzoyl] -4-piperidyl]benzonitrile
Figure imgf000040_0002
A mixture of 3-amino-4-trifluoromethoxy benzoic acid (Step 2) (2.8 g, 12.66 mmol), 4-(4'- cyanophenyl)piperidine (2.36 g, 12.66 mmol, 1 eq), N-(3-Dimethylaminopropyi)-N'- ethylcarbodiimide hydrochloride (EDAC) (2.67 g, 13.93 mmol, 1.1 eq) and DMAP (155 mg, 1.27 mmol, 0.1 eq) in DMF (30 mL) was stirred at room temperature for 2 hrs. EtOAc (200 mL) was added and the resulting mixture washed sequentially with aqueous potassium bisulfate solution (100 mL of 2M KHSO4), brine (100 mL), dried (MgSO4), filtered and reduced in vacuo to give a brown oil. Ethyl acetate was added and the resulting colourless solid isolated by filtration. A precipitate also appeared during the extraction process; this was isolated and the solids combined to give the title compound (3.16 g), ^H NMR (300.072 MHz, CDCl3) δ 1.43 - 2.02 (4H, m),2.77 - 3.22 (3H, m),3.80 - 4.21 (3H, m),4.63 - 5.03 (IH, m),6.72 - 6.77 (IH, m),6.87 (IH, d),7.13 - 7.18 (IH, m),7.32 (2H, d),7.61 (2H, d), m/z 390 (M+H)+. Intermediate D 4- [ 1 -(5 -amino-2-methyl-benzoyl)-4-piperidyl]benzonitrile
Figure imgf000040_0003
4-[l-(2-methyl-5-nitro-benzoyl)-4-piperidyl]benzonitrile
Figure imgf000041_0001
A mixture of 2-methyl-5-nitrobenzoic acid (5g, 27.6mmol), 4-(4'-cyanophenyl)piperidine (5.14g, 27.6mmol), N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (5.82g, 30.36mmol) and DMAP (338mg, 2.76 mmol) in DMF (50 mL) was stirred at room temperature for 2 hrs. Ethyl acetate (200ml) was added and the resulting solution washed with dilute hydrochloric acid (100 mL of IM), NaHCO3, brine (100 mL). At this point a colourless solid precipitated, which was isolated by filtration and dried . The filtrate was dried (MgSO4), filtered and reduced in vacuo to give a white solid. This was chromatographed (12O g silica column, Companion, eluting with a gradient consisting of 0- 50% ethyl acetate in isohexane) to give a colourless solid which was identical to that isolated previously. The solids were combined to give the title compound (4 g), 1H NMR (300.072 MHz, CDCl3) δl.64 - 2.11 (4H, m),2.42 - 2.53 (3H, m),2.79 - 3.00 (2H, m),3.09 - 3.23 (IH, m),3.55 (IH, d),4.98 (IH, d),7.29 - 7.36 (2H, m),7.42 (IH, d),7.63 (2H, d),8.04 - 8.18 (2H, m), m/z 348 (M-H)-.
Step 2; 4-[ 1 -(5-amino-2-methyl-benzoyi)-4-piperidyi]benzonitrile
Figure imgf000041_0002
A solution of 4-[l-(2-methyl-5-nitro-benzoyl)-4-piperidyl]benzonitrile (4g, 11.45 mmol) in ethanol / THF (200 mL of a 1 : 1 mixture) was placed under an atmosphere of argon and treated with 10% palladium on carbon catalyst (0.6 g, 15% by weight). The reaction mixture was stirred under a hydrogen atmosphere for 4hrs. The catalyst was removed by filtration through celite, and the filtrate evaporated in vacuo to give a yellow foam. Some starting material still remained so the hydrogenation was repeated as above, with stirring for a further 1 hr. The isolation was repeated as above to give a yellow foam. EtOAc was added and the solution washed with water and brine; the solvent was removed in vacuo to give the title compound as a yellow solid (3.3g), 1H NMR (400.132 MHz, DMSO) δ 1.39 - 1.62 (2H, m),1.69 - 1.78 (IH, m),1.88 (IH, d),1.99 - 2.12 (3H, m),2.80 (IH, t),2.90 - 2.99 (IH, m),3.08 (IH, t),3.45 - 3.51 (IH, m),4.63 - 4.72 (IH, m),5.01 (2H, s),6.37 (IH, d),6.48 - 6.52 (IH, m),6.89 (IH, d),7.46 - 7.52 (2H, m),7.78 (2H, d), m/z 320 (M+H)+. Intermediate E 4-[ 1 -(3-amino-4-fluoro-benzoyl)-4-piperidyl]benzonitrile
Figure imgf000042_0001
The title compound was prepared in a manner similar to that described for Intermediate A, starting from 3-amino-4-fluoro-benzoic acid and 4-(4'-cyanophenyl)piperidine, 1H NMR
(300.073 MHz, dmso) δ 1.43 - 2.00 (m, 4H), 2.68 - 3.23 (m, 3H), 3.59 - 4.05 (m, IH),
4.21 - 4.88 (m, IH), 5.29 (s, 2H), 6.49 - 6.61 (m, IH), 6.80 (d, IH), 6.95 - 7.08 (m, IH),
7.49 (d, 2H), 7.76 (d, 2H), m/z 324 (M+H)+.
Intermediate F
4-[ 1 -(5-amino-2,4-dimethyl-benzoyl)-4-piperidyl]benzonitrile
Figure imgf000042_0002
4-[l-(2,4-dimethyl-5-nitro-benzoyl)-4-piperidyl]benzonitrile
Figure imgf000042_0003
The title compound was prepared in a manner similar to that described for Intermediate C,
Step 1, starting from 2,4-dimethyl-5-nitro-benzoic acid and 4-(4'-cyanophenyl)piperidine;
1H NMR (300.072 MHz, CDCl3) δ 1.44 - 1.64 (m, IH), 1.66 - 1.91 (m, 2H), 1.95 - 2.09
(m, IH) ,2.30 - 2.49 (br s, 3H), 2.61 (s, 3H), 2.79 - 2.95 (m, 2H), 3.05 - 3.26 (m, IH), 3.59
(d, IH), 4.95 (d, IH), 7.22 (s, IH), 7.32 (d, 2H), 7.61 (d, 2H), 7.81 (br s, IH), m/z 405
(M+MeCN+H)+.
Step 2: 4- [ 1 -(5 -amino-2,4-dimethyl-benzoyl)-4-piperidyl]benzonitrile
Figure imgf000043_0001
The title compound was prepared in a manner similar to that described for Intermediate C, Step 2, starting from 4-[l-(2,4-dimethyl-5-nitro-benzoyl)-4-piperidyl]benzonitrile, and using a methanol / THF mixture (1:1) as solvent; 1H NMR (300.073 MHz, dmso) δ 1.36 - 1.63 (m, 2H), 1.64 - 1.79 (m, IH), 1.80 - 1.95 (m, IH), 1.96 - 2.08 (br s, 3H), 2.02 (s, 3H),
2.69 - 2.85 (m, IH), 2.85 - 2.97 (m, IH), 2.98 - 3.12 (m, IH), 3.40 - 3.56 (m, IH), 4.59 -
4.70 (m, IH), 4.73 (br s, 2H), 6.30 - 6.55 (br m, IH), 6.78 (s, IH), 7.47 (d, 2H), 7.77 (d, 2H); peak broadening is observed due to conformations of amide group, m/z 334 (M+H)+. Intermediate G o 4-[l-(3-amino-4-methoxy-benzoyl)-4-piperidyl]benzonitrile
Figure imgf000043_0002
A stirred mixture of 4-(4'-cyanophenyl)piperidine (3 g, 16 mmol), 3-amino-4- methoxybenzoic acid (2.675 g, 16 mmol, 1 eq) and DIPEA (4.2 ml, 24 mmol, 1.5 eq) in DCM (100 mL) was blanketed with nitrogen and treated with N-(3-Dimethylaminopropyl)-5 N'-ethylcarbodiimide hydrochloride (EDAC) (3.4 g, 17.6 mmol, 1.1 eq). The reaction mixture was stirred for three days. Addition of water to the reaction mixture resulted in an emulsion. Evaporation of most of the DCM resulted in the formation of a solid precipitate. The solid was isolated by filtration and washed with EtOAc (2 x 75 mL portions) to give a colourless solid (2.5 g). The ethyl acetate washings were combined and added to theo aqueous filtrate; the phases were separated and the organic phase washed with water, dried (MgSO4) and evaporated to give a further 2 g of colourless solid. The solids thus prepared were identical and combined to give the title compound as (4.5 g, 83%), 1H NMR (300.073 MHz, dmso, 30°C) δ 1.48 - 1.67 (2H, m), 1.71 - 1.87 (2H, m), 2.80 - 3.08 (3H, m), 3.78 (3H, s), 3.88 - 4.63 (2H, m), 4.84 (2H, s), 6.56 - 6.64 (IH, m), 6.67 - 6.73 (IH, "5 m), 6.80 (IH, dJ = 8.1 Hz), 7.49 (2H, dJ = 8.1 Hz), 7.76 (2H, dJ = 9.4 Hz), m/z 336 (M+H)+. Intermediate H
4-[l-(3-amino-4-methyl-benzoyl)-4-piperidyl]-N,N-dimethyl-benzamide
Figure imgf000044_0001
Step 1: Methyl 4-[l-(4-methyl-3-nitro-benzoyl)-4-piperidyl]benzoate
Figure imgf000044_0002
A solution of 4-methyl-3-nitrobenzoyl chloride (3.9 g, 19.55 mmol) in DCM (50 niL) was added dropwise to a solution of methyl 4-(4-piperidyl)benzoate (5 g, 19.55 mmol) and DIPEA in DCM (100 mL). The reaction mixture was stirred at ambient temperature for 72 hrs. The reaction mixture was then washed sequentially with saturated aqueous sodium hydrogen carbonate solution, IM aqueous citric acid and water. The solvent was dried (phase separating cartridge) and evaporated to give the title compound as a brown waxy solid (6.85 g, 92%), 1H NMR (300.073 MHz, DMSOd6) δl.56-1.95 (4H, m, 2.56 (3H, s), 2.94 (2H, t), 3.60 - 3.67 (IH, m), 3.86 (3H, s), 4.64 (IH, s), 7.46 (2H, d), 7.60 (IH, d), 7.71 - 7.74 (IH, m), 7.92 (2H, d), 8.05 (IH, d), m/z 383 (M+H)+. Step 2: 4- [ 1 -(4-methyl-3 -nitro-benzoyl)-4-piperidyl]benzoic acid
A suspension of methyl 4-[l-(4-methyl-3-nitro-benzoyl)-4-piρeridyl]benzoate (Step 1) (5.68 g, 14.9 mmol) in MeOH (57 mL) was treated with aqueous sodium hydroxide solution (19 mL of 2M, 37.1 mmol, 2 eq.) and the reaction mixture stirred at 5O0C for two hrs. More MeOH (25 mL) was added and stirring for continued for one hr. The reaction mixture was cooled and treated with 2M aqueous hydrochloric acid to <pH5, diluted with EtOAc, and the organic layer separated. The aqueous portion was shaken with more EtOAc and the organic layer again separated. The organic extracts were combined, washed with brine, dried over MgSO4, filtered and evaporated to give the title compound as a yellow solid (4.92 g), 1H NMR (300.073 MHz, dmso) δ 1.56 - 1.77 (m, 4H), 2.54 (s, 3H), 2.68 - 3.00 (m, 3H), 3.51 - 3.73 (m, IH), 4.52 - 4.71 (m, IH), 7.41 (d, J= 8.3 Hz, 2H), 7.58 (d, J= 7.9 Hz, IH), 7.70 (d, J= 9.4 Hz, IH), 7.87 (d, J= 8.2 Hz, 2H), 8.02 (s, IH), 12.69 - 12.99 (m, IH), m/z 367 (M-H)". Step 3: N,N-dimethyl-4-[l-(4-methyl-3-nitro-benzoyl)-4-piperidyl]benzamide
Figure imgf000045_0001
A solution of 4-[l-(4-methyl-3-nitro-benzoyl)-4-piperidyl]benzoic acid (Step 2) (2.89 g, 7.84 mmol), dimethylamine (5.89 mL of a 2M solution in THF, 11.8 mmol, 1.5 eq.), DIPEA (2.7 mL, 15.7 mmol, 2 eq.), and DMAP (2.1 g, 17.3.mmol, 2.2 eq.) in DCM (58 mL) was treated with EDAC (1.8 g, 9.4 mmol, 1.2 eq.), and the reaction mixture stirred for 16 hrs at ambient temperature. Further reagents were added and the reaction mixture stirred for a further 16 hr by which time reaction was essentially complete. The reaction mixture was washed with water and the phases separated. The organic portion was concentrated to a brown solid. The crude product was re-dissolved in DCM and the solution washed sequentially with water, citric acid solution (IM in water) and saturated sodium bicarbonate solution. The organic phase was dried (MgSO4), filtered and concentrated to a brown solid, which was purified by chromatography (12Og silica column, gradient eluting with 10 - 50% EtOAc in iso-hexane) to give the title compound as a yellow gum, (1.74 g), 1H NMR (300.073 MHz, dmso) δ 1.57 - 1.92 (m, 4H), 2.54 (s, 3H), 2.76 - 3.03 (m, 9H), 3.57 - 3.71 (m, IH), 4.51 - 4.70 (m, IH), 7.34 (s, 4H), 7.58 (d, J= 7.9 Hz, IH), 7.70 (d, J = 6.2 Hz, IH), 8.02 (s, IH), m/z 396 (M+H)+. Step 4: 4-[l-(3-amino-4-methyl-benzoyl)-4-piperidyl]-N,N-dimethyl-benzamide
Figure imgf000046_0001
A solution of N,N-dimethyl-4-[l-(4-methyl-3-nitro-benzoyl)-4-piperidyl] benzamide (Step 3) (1.74g) in MeOH (35 tnL) was treated with palladium-on-charcoal catalyst (122 mg of 10% Pd/C). The reaction mixture was stirred in an atmosphere of hydrogen at ambient temperature and pressure for 2 hours. The catalyst was removed by filtration and the filtrate concentrated to give the title compound as a colourless solid (1.5Ig), 1H NMR (300.073 MHz, dmso) δ 1.46 - 1.68 (m, 2H), 1.69 - 1.88 (m, 2H), 2.06 (s, 3H), 2.74 - 3.07 (m, 9H), 3.64 - 3.97 (m, IH), 4.43 - 4.70 (m, IH), 4.97 (s, 2H), 6.49 (d, J= 7.4 Hz, IH), 6.64 (s, IH), 6.95 (d, J= 7.5 Hz, IH), 7.32 (s, 4H), m/z 366 (M+H)+. Intermediate I 4-[l-(3-amino-4-methyl-benzoyl)-4-piperidyl]benzamide
Figure imgf000046_0002
Step 1:
4-[l-(4-methyl-3-nitro-benzoyl)-4-piperidyl]benzamide
Figure imgf000046_0003
The title compound was prepared from 4-[l-(4-methyl-3-nitro-benzoyl)-4-piperidyl] benzoic acid (Intermediate H, Step 2) using the procedure described in Heterocycles, 2006, 68 (6), 1149-1162, 1H NMR (300.073 MHz, dmso) δ 1.57 - 1.94 (m, 4H), 2.54 (s, 3H), 2.66 - 3.03 (m, 3H), 3.56 (s, 3H), 3.59 - 3.72 (m, IH), 4.50 - 4.71 (m, IH), 7.24 (s, IH), 7.35 (d, J= 8.1 Hz, 2H), 7.57 (d, J= 7.9 Hz, IH), 7.70 (d, J= 7.8 Hz, IH), 7.80 (d, J= 8.1 Hz, 2H), 7.87 (s, IH), 8.02 (s, IH), m/z 368 (M+H)+. Step 2:
4-[ 1 -(3-amino-4-methyl-benzoyl)-4-piperidyl]benzamide
Figure imgf000047_0001
The title compound was prepared from 4-[l-(4-methyl-3-nitro-benzoyl)-4-piperidyl] benzamide (Step 1) using the hydrogenation procedure described in Intermediate H, Step 4, 1H NMR (300.073 MHz, dmso) δ 1.47 - 1.67 (m, 2H), 1.70 - 1.88 (m, 2H), 2.06 (s, 3H), 2.69 - 2.98 (m, 3H), 3.66 - 3.98 (m, IH), 4.37 - 4.74 (m, IH), 4.97 (s, 2H), 6.49 (d, J = 7.5 Hz, IH), 6.64 (s, IH), 6.95 (d, J= 7.5 Hz, IH), 7.24 (s, IH), 7.33 (d, J= 8.2 Hz, 2H), 7.80 (d, J= 8.1 Hz, 2H), 7.87 (s, IH), m/z 368 (M+H)+.

Claims

Claims
1) A compound of formula I
Figure imgf000048_0001
or a pharmaceutically acceptable salt thereof, in which
R1 represents 1) a
Figure imgf000048_0002
group optionally substituted by one or two groups selected from A-S below and/or by one to five groups selected from T below: A) phenyl optionally substituted by one or more of the following i) halo; ii) cyano; iii) a C1-4alkoxy group optionally substituted by one or more halo iv) hydroxy; v) a Ci-4alkyl group optionally substituted by one or more halo; vi) carbamoyl; vii) N-C1-
6alkylcarbamoyl; viii)
Figure imgf000048_0003
; ix) carboxy; x) C1-6 alkoxycarbonyl; xi) Ci-βalkylthio; xii) Ci-βalkylsulfinyl; xiii) Ci-βalkylsulfonyl; xiv) Ci-6alkylsulfonyloxy; xv) sulphamoyl; xvi) N-Ci-βalkylsulphamoyl; xvii) N, N-diCi-βalkylsulphamoyl; xviii) benzyl xix) benzyloxy; xx) heteroaryl; xxi) heteroaryloxy; xxii) phenyl xxiii) phenoxy xxiv) phenylsulphamoyl; xxv) heteroarylsulphamoyl; xxvi) a carbon linked saturated or partially unsaturated 4 to 8 membered heterocyclic group as defined in c) below; xxvii) phenylsulfoiiyl ; xxviii) heteroarylsulfonyl; xxix) a group of formula ΝRcRd in which Rc and Rd independently represent: a) H; b) Cμβalkanoyl; c) a carbon linked saturated or partially unsaturated 4 to 8 membered heterocyclic group containing one or more N, S or O, wherein the S may be in its oxidised form of SO or SO2 , which is optionally fused to a benz ring and any ring is optionally substituted by one or more of the following: hydroxy, oxo, carboxy, a C1-6alkoxy group optionally substituted by one or more hydroxy or Ci^alkoxy, Ci-4alkanoyl, benzoyl, amino, C1-3alkylamino, di(C1-3 alkyl)amino or a Ci^alkyl optionally substituted by one or more hydroxy or C^aU-oxy; d) a Ci^alkyl group optionally substituted by one or more of the following: hydroxy; carboxy; a Cμδalkoxycarbonyl group; a Ci-βalkoxy group; heteroaryl; a group of formula NReRf in which Re and Rf independently represent H; a Ci^alkanoyl group; a C1. 6alkylsulphonyl group; a Ci-όalkoxycarbonyl group; a C^alkyl group optionally substituted by one or more hydroxy or Ci^alkoxy , or Re and Rf together with the nitrogen atom to which they are attached represent a saturated or partially unsaturated 4 to 8 membered heterocyclic ring optionally containing an additional sulphur including oxidised as SO or SO2 , oxygen or nitrogen and/or optionally fused to a benz ring and any ring is optionally substituted by one or more of the following: a Ci-βalkoxy group; carboxy; a C1. 6alkylsulfonyl group; Ci-4alkanoyl; benzoyl; hydroxy; oxo; carboxy; or a Ci,6alkyl group optionally substituted by one or more hydroxy or by one or more C^alkoxy or by one or more carboxy; e) Rc and Rd together with the nitrogen atom to which they are attached represent a saturated or partially unsaturated 4 to 8 membered heterocyclic ring optionally containing an additional oxygen, sulphur, SO, SO2 or nitrogen and/or optionally fused to a benz ring and/or optionally substituted by one or more of the following: a Ci-6alkoxy group; C1. 4alkanoylgroup; benzoyl; a Ci-6alkoxycarbonyl group; a Ci-βalkylsulfonyl group; carbamoyl; N-d-δalkylcarbamoyl; N, N-diC1-6alkylcarbamoyl; hydroxy; oxo; carboxy; a C^aUcyl group (which is optionally substituted by one or more of the following: a C1. 6alkoxy group, hydroxy or a group of formula ΝReRf in which Re and Rf are as defined above) or a group of formula NReRf in which Re and Rf are as defined above; f) a Ci-βalkylsulphonyl group; g) phenylsulfonyl; h) heteroarylsulfonyl; i) benzoyl; j) phenyl optionally substituted by one or more of the following: halo; C^alkyl; C1-
3alkoxy; a C1-6alkanoylamino group; carbamoyl; N-Ci-6alkylcarbamoyl; N,N-did.
6alkylcarbamoyl; k) heteroaryl optionally substituted by one or more carboxy; fluoro; hydroxy; a Ci-3alkoxy group optionally substituted on C2 or C3 by carboxy; a group ΝRcRd in which Rc and Rd are as defined above; or a group CONReRf in which Re and Rf are as defined above; 1) a C3-iocycloalkyl group which may be monocyclic, bicyclic or tricyclic and optionally may be bridged and is optionally substituted by one or more carboxy; fluoro; hydroxy; a Ci-3alkoxy group optionally substituted on C2 or C3 by carboxy; a group NReRf in which Re and Rf are as defined above; or a group CONReRf in which Re and Rf are as defined above; m) a Ci-6alkoxycarbonyl group;
B) a heteroaryl group which is optionally substituted by groups i) to xxix) as described for phenyl above;
C) a group of formula NRcRd in which Rc and Rd are as defined above; o D) a C3-7cycloalkyl group optionally substituted by one or more hydroxy or a group of formula NReRf in which Re and Rf are as defined above;
E) a carbon linked saturated or partially unsaturated 4 to 8 membered heterocyclic group containing one or more N, S or O, wherein the S may be in its oxidised form of SO or SO2, which is optionally fused to a benz ring and/or is optionally substituted by one ors more of the following: hydroxy; oxo; a C1-6alkoxy group; carboxy; hydroxy; Ci-4alkanoyl; a C1-6alkylsulfonyl group; amino; C1-3alkylamino; di(C1-3 alkyl)amino; or a Ci-βalkyl optionally substituted by one or more hydroxy or C1-6alkoxy;
F) a C1-6 alkoxycarbonyl group;
G) a C2-6alkynyl group: Q H) a group -CONRcRd in which Rc and Rd are as defined above;
I) a Ci_6alkoxy group;
J) a C2-6alkenyl group:
K) a Ci-6alkyl group;
L) a C1-6alkylsulphonyl group; 5 M) phenylsulfonyl;
N) heteroarylsulfonyl;
O) benzoyl;
P) a C^alkanoyl group
Q) hydroxy; 0 R) oxo;
S) carboxy;
T) fluoro or R1 represents
2) a C3-7cycloalkyl group optionally substituted by one or two groups selected from A to T above;
3) a C2-6alkynyl group optionally substituted by one or two groups selected from A to T above;
4) a carbon linked saturated or partially unsaturated 4 to 8 membered heterocyclic group containing one or more N, S or O, wherein the S may be in its oxidised form of SO or SO2, which is optionally fused to a benz ring and any ring is optionally substituted by a group A to T as defined above one or more of the following: hydroxy, oxo, carboxy, a Ci-6alkoxy group, hydroxy, a Ci-βalkylsulfonyl group, C1-4alkanoyl, benzoyl, amino, C1- 3alkylamino, di(Ci-3 alkyl)amino or a Ci^alkyl optionally substituted by one or more hydroxy or Ci^alkoxy;
5) a C2-6alkenyl group optionally substituted by one or two groups selected from A to T above; 6) optionally substituted phenyl including optional fusion of the phenyl ring to a saturated or partially unsaturated 5 to 6 membered heterocyclic ring optionally containing one, two or three hetero atoms selected from oxygen, sulphur optionally in its oxidised forms of SO or SO2 or nitrogen wherein the heterocyclic ring is optionally substituted by one or more of the following: a Ci-βalkoxy group; a C^alkanoyl group; carboxy; a Ci.6alkylsulfonyl group; a C^aUcoxycarbonyl group; carbamoyl; N-Ci-βalkylcarbamoyl; N, N-diCμ
6alkylcarbamoyl; hydroxy; oxo; a C^alkyl group (which is optionally substituted by one or more of the following: a Ci^alkoxy group, hydroxy or a group of formula ΝR°Rd in which Rc and Rd are as defined above) and wherein the phenyl ring is optionally substituted by one or more of the groups i to xxix listed above or by a heteroaryl group optionally substituted by one or more groups i) to xxix) above or by an ureido group of formula RmRnN-C(O)-NH- in which Rm and Rn independently represent H, a C1-6alkyl group optionally substituted by a C^alkoxy group, or Rm and Rn together with the nitrogen atom to which they are attached represent a saturated or partially unsaturated 4 to 8 membered heterocyclic ring optionally containing an additional sulphur including oxidised as SO or SO2 , oxygen or nitrogen and/or optionally fused to a benz ring and/or optionally substituted by one or more of the following: a d-βalkoxy group; hydroxy; oxo; carboxy; a Q-βalkylsulfonyl group; or a C^ealkyl group optionally substituted by one or more hydroxy or Ci-6alkoxy;
7) optionally substituted heteroaryl including N-oxides and S-oxides thereof optionally substituted by one or more of the groups i to xxi listed above; wherein any alkyl chain mentioned in any of the definitions from A to P above or in any of the definitions i to xxix above is optionally substituted by 1) one group selected from: carboxy; hydroxy; a C1-3alkoxy group optionally substituted on C2 or C3 by carboxy; a group NRcRd in which R° and Rd are as defined above; or a group CONReRf in which Re and Rf are as defined above; and /or by 2) from one to five fluoro; and further wherein any cycloalkyl, phenyl, heteroaryl ring or carbon linked saturated or partially saturated 4 to 8 membered heterocyclic group in the list of optional substituents from A to P above or in any of the definitions i to xxix above, for which specific substitution has not been previously mentioned, is optionally substituted by one group selected from: carboxy; hydroxy; a C1-3alkoxy group optionally substituted on C2 or C3 by carboxy; a group NRcRd in which Rc and Rd are as defined above; or a group CONReRf in which Re and Rf are as defined above; and /or is optionally substituted by one to five fluoro;
R2 represents H, halo, a C1-3alkyl group, trifluoromethoxy or cyano; R3 represents H, halo, a Ci-3alkyl group, trifluoromethoxy or cyano; R4 represents i) H, ii) a C1-3alkyl group optionally substituted by one or more halo iii) a C1- 3alkoxy group optionally substituted by one or more halo iv) halo, v) nitro, vi) cyano, vii) a Chalky IS (O)y(O)z- wherein y is 0,1 or 2 and z is 0 except when y is 2 when z is 0 or 1 viii) a group CH2NRURV in which Ru and Rv independently represent H; a Ci-3alkylsulphonyl group, a C^alkanoyl group or a C1-3alkyl group or Ru and Rv together with the nitrogen atom to which they are attached represent azetidinyl, pyrrolidinyl, piperidinyl or morpholinyl; ix) a group CO2RW in which Rw is a Ci^alkyl group; or x) a group CONRxRy in which Rx and Ry independently represent H; or a Ci-3alkyl group or Rx and Ry together with the nitrogen atom to which they are attached represent azetidinyl; pyrrolidinyl, piperidinyl or morpholinyl; R5 and R5' independently represent H, halo, cyano, C1-3alkyl optionally substituted by one or more halo or C1-3alkoxy optionally substituted by one or more halo; R6 and R6 independently represent H, halo, cyano, Ci-3alkyl optionally substituted by one or more halo or Ci-3alkoxy optionally substituted by one or more halo; and R7 is H or OH but excluding the following compounds: tert-butyl N-[3-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]phenyl]carbamate 2-methylpropyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2- (trifluoromethoxy)phenyl]carbamate
2-methylpropyl N-[2-methyl-5-[4-(4-methylsulfonylphenyl)piperidine-l- carbonyl]phenyl]carbamate and 2-methylpropyl N-[3-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-4-fluorophenyl]carbamate.
2) A compound as claimed in claim 1 in which R7 is H.
3) A compound as claimed in claim 1 as represented by formula II
Figure imgf000053_0001
or a pharmaceutically acceptable salt thereof, in which
R1 represents 1) a Cμβalkyl group optionally substituted by one or more of the following: a) phenyl b) pyridyl or c) a Ci^alkoxy group 2) a C3-7cycloalkyl group or 3) phenyl ;
R2 represents H, halo, a C^alkyl group; or trifiuoromethoxy;
R3 represents H, halo, a C^alkyl group; or trifiuoromethoxy; provided that one of R2 and R3 is other than H; and R4 represents cyano.
4) A compound according to any previous claim in which R1 represents a Cμealkyl group optionally substituted by 3-pyridyl or 4-pyridyl.
5) A compound according to any previous claim in which R2 represents methyl and R3 is H. 6) A compound selected from one or more of the following compounds:
2-methylpropyl N-[5~[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl- phenyl]carbamate; propan-2-yl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate benzyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate; phenyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate; benzyl N-[2-chloro-5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]phenyl]carbamate; benzyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-(trifluoromethoxy)phenyl]- carbamate; Q benzyl N-[3-[4-(4-cyanophenyl)piperidine-l-carbonyl]-4-methyl-phenyl]carbamate; benzyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-fluoro-phenyl]carbamate; benzyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2,4-dimethyl-phenyl]carbamate;
2-methylpropyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2,4-dimethyl- phenyl]carbamate; s pyridin-3-ylmethyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl- phenyl]carbamate; pyridin-4-ylmethylN-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl- phenyl]carbamate; methyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate;Q 2-methoxyethyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl- phenyljcarbamate; phenethyl N- [5 - [4-(4-cyanopheny l)piperidine- 1 -carbonyl] -2-methyl-phenyl] carbamate ethyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate; propyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methyl-phenyl]carbamate;5 benzyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-methoxy-phenyl]carbamate; tert-Butyl N-[5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate; benzyl N-[5-[4-[4-(dimethylcarbamoyl)phenyl]piperidine-l-carbonyl]-2-methyl- phenyl]carbamate; benzyl N-[5-[4-(4-carbamoylphenyl)piperidine- 1 -carbonyl]-2-methyl-phenyl]carbamate;0 2-methylpropyl N-[2-chloro-5-[4-(4-cyanophenyl)piρeridine-l-carbonyl]ρhenyl]- carbamate; phenyl N-[2-chloro-5-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]phenyl]carbamate; 2-methylpropyl N-[3-[4-(4-cyanophenyl)piperidine- 1 -carbonyl]-4-methylphenyl]- carbamate;
2-methylpropyl N-[5-[4-(4-cyanophenyl)piperidine-l-carbonyl]-2-fluorophenyl]carbamate; 2-methylpropyl N-[3-[4-(4-cyanophenyl)piperidine- 1 -carbonyljphenyljcarbamate; and benzyl N-[3-[4-(4-cyanophenyl)piperidine-l-carbonyl]phenyl]carbamate or a pharmaceutically-acceptable salt thereof.
7) A method of treating obesity or being overweight, eating disorders, dyslipidaemia and type 2 diabetes mellitus comprising administering a pharmacologically effective amount of a compound of formula I as defined in any one of claims 1 to 6 to a patient in need thereof.
8) A pharmaceutical formulation comprising a compound of formula I as claimed in any one of claims 1 to 6 in admixture with pharmaceutically acceptable adjuvants, diluents and/or carriers. 9) A process for preparing a compound of formula I as claimed in claim 1 comprising:, (a) reacting a compound of formula VI
Figure imgf000055_0001
with a compound of formula VII R1OC(O)X
VII wherein X is a leaving group in the presence of a diluent optionally in presence of a base at a temperature in the range of 0-1500C;
b) reacting a compound of formula VIII
Figure imgf000056_0001
VIIl with a compound of formula IX
Figure imgf000056_0002
IX in which X represents a leaving group in the presence of a diluent and optionally in the presence of a base at a temperature in the range of 0-15O0C; or c) reacting a compound of formula VIII
Figure imgf000056_0003
VIII with a compound of formula X
Figure imgf000056_0004
X in which X represents a replaceable group in the presence of carbon monoxide and in the presence of a metal catalyst in a solvent at a temperature in the range 0 - 150°C; or d) reacting a compound of formula VIII
Figure imgf000057_0001
VIII with a compound of formula XI
Figure imgf000057_0002
Xl optionally in the presence of a coupling agent and optionally in the presence of a diluent at a temperature in the range of 0-1500C; wherein R1, R2, R3, R4, R5, R5', R6, R6, and R7 are as defined in claim 1
10) A compound of formula VI as defined in claim 9.
PCT/GB2007/004936 2006-12-21 2007-12-20 Piperidine derivatives for the treatment of obesity WO2008075077A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US87118706P 2006-12-21 2006-12-21
US60/871,187 2006-12-21
US90870607P 2007-03-29 2007-03-29
US60/908,706 2007-03-29

Publications (1)

Publication Number Publication Date
WO2008075077A1 true WO2008075077A1 (en) 2008-06-26

Family

ID=39110735

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2007/004936 WO2008075077A1 (en) 2006-12-21 2007-12-20 Piperidine derivatives for the treatment of obesity

Country Status (1)

Country Link
WO (1) WO2008075077A1 (en)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120264737A1 (en) * 2011-03-08 2012-10-18 3-V Biosciences, Inc. Heterocyclic Modulators of Lipid Synthesis
WO2013022927A2 (en) * 2011-08-08 2013-02-14 The Brigham And Women's Hospital, Inc. Treatment of uterine leiomyomata
WO2014008197A1 (en) * 2012-07-03 2014-01-09 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
WO2015095767A1 (en) * 2013-12-20 2015-06-25 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis and combinations thereof
WO2015105860A1 (en) * 2014-01-07 2015-07-16 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis for use against cancer and viral infections
AU2012225390B2 (en) * 2011-03-08 2015-11-12 Sagimet Biosciences Inc. Heterocyclic modulators of lipid synthesis
US9624173B2 (en) 2011-03-08 2017-04-18 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
US10189822B2 (en) 2015-03-19 2019-01-29 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
US10363249B2 (en) 2014-08-15 2019-07-30 3-V Biosciences, Inc. Fatty acid synthase inhibitor for use in the treatment of drug resistant cancer
US10399951B2 (en) 2013-03-13 2019-09-03 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10793554B2 (en) 2018-10-29 2020-10-06 Forma Therapeutics, Inc. Solid forms of 4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl)piperazin-1-yl)(1-hydroxycyclopropyl)methanone
US10875848B2 (en) 2018-10-10 2020-12-29 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US11034690B2 (en) 2016-11-11 2021-06-15 Saginiet Biosciences Inc. Heterocyclic modulators of lipid synthesis
WO2021130723A1 (en) * 2019-12-24 2021-07-01 Cadila Healthcare Limited Novel compounds suitable for the treatment of dyslipidemia
RU2774758C2 (en) * 2015-03-19 2022-06-22 Сагимет Байосайенсиз Инк. Heterocyclic lipid synthesis modulators
US11622968B2 (en) 2011-03-08 2023-04-11 Sagimet Biosciences Inc. Heterocyclic modulators of lipid synthesis

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5981575A (en) * 1996-11-15 1999-11-09 Johns Hopkins University, The Inhibition of fatty acid synthase as a means to reduce adipocyte mass
WO2001090101A1 (en) * 2000-05-22 2001-11-29 Aventis Pharmaceuticals Inc. Arylmethylamine derivatives for use as tryptase inhibitors
WO2002055661A2 (en) * 2000-11-17 2002-07-18 Smithkline Beecham Corporation Fatty acid synthase inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5981575A (en) * 1996-11-15 1999-11-09 Johns Hopkins University, The Inhibition of fatty acid synthase as a means to reduce adipocyte mass
WO2001090101A1 (en) * 2000-05-22 2001-11-29 Aventis Pharmaceuticals Inc. Arylmethylamine derivatives for use as tryptase inhibitors
WO2002055661A2 (en) * 2000-11-17 2002-07-18 Smithkline Beecham Corporation Fatty acid synthase inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
V. K. AGRAWAL ET. AL.: "Antiparasitic Agents: Part VI. Synthesis of 7-chloro-4-(4-substituted phenylamino)- & 7-Chloro-4-(4-substituted-piperazin-1-yl)-quinolines as Potential Antiparasitic Agents.", INDIAN JOURNAL OF CHEMISTRY, vol. 26B, 1 June 1987 (1987-06-01), pages 550 - 555, XP002919361 *

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120264737A1 (en) * 2011-03-08 2012-10-18 3-V Biosciences, Inc. Heterocyclic Modulators of Lipid Synthesis
US9624173B2 (en) 2011-03-08 2017-04-18 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
US9809591B2 (en) 2011-03-08 2017-11-07 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
AU2012225390B2 (en) * 2011-03-08 2015-11-12 Sagimet Biosciences Inc. Heterocyclic modulators of lipid synthesis
US8871790B2 (en) * 2011-03-08 2014-10-28 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
US11622968B2 (en) 2011-03-08 2023-04-11 Sagimet Biosciences Inc. Heterocyclic modulators of lipid synthesis
WO2013022927A2 (en) * 2011-08-08 2013-02-14 The Brigham And Women's Hospital, Inc. Treatment of uterine leiomyomata
WO2013022927A3 (en) * 2011-08-08 2013-04-18 The Brigham And Women's Hospital, Inc. Treatment of uterine leiomyomata
KR20150032571A (en) * 2012-07-03 2015-03-26 3-브이 바이오사이언시스, 인코포레이티드 Heterocyclic modulators of lipid synthesis
CN104507928A (en) * 2012-07-03 2015-04-08 3-V生物科学股份有限公司 Heterocyclic modulators of lipid synthesis
WO2014008197A1 (en) * 2012-07-03 2014-01-09 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
US9428502B2 (en) 2012-07-03 2016-08-30 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
JP2015522036A (en) * 2012-07-03 2015-08-03 3−ブイ・バイオサイエンシーズ・インコーポレイテッド3−V Biosciences,Inc. Heterocyclic modulator of lipid synthesis
CN104507928B (en) * 2012-07-03 2016-09-28 3-V生物科学股份有限公司 The heterocyclic modulators of lipid synthesis
KR102071095B1 (en) 2012-07-03 2020-01-29 3-브이 바이오사이언시스, 인코포레이티드 Heterocyclic modulators of lipid synthesis
US10118913B2 (en) 2012-07-03 2018-11-06 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
US10053450B2 (en) 2012-07-03 2018-08-21 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
RU2779069C2 (en) * 2012-07-03 2022-08-31 Сагимет Байосайенсиз Инк. Heterocyclic lipid synthesis modulators
US10800750B2 (en) 2013-03-13 2020-10-13 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10399951B2 (en) 2013-03-13 2019-09-03 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10995078B2 (en) 2013-03-13 2021-05-04 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10472342B2 (en) 2013-03-13 2019-11-12 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10457655B2 (en) 2013-03-13 2019-10-29 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10450286B2 (en) 2013-03-13 2019-10-22 Forma Therapeutics, Inc. Compounds and compositions for inhibition of FASN
US10226449B2 (en) 2013-12-20 2019-03-12 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis and combinations thereof
CN106061963B (en) * 2013-12-20 2019-10-22 3-V生物科学股份有限公司 The heterocyclic modulators and a combination thereof of lipid synthesis
WO2015095767A1 (en) * 2013-12-20 2015-06-25 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis and combinations thereof
CN106061963A (en) * 2013-12-20 2016-10-26 3-V生物科学股份有限公司 Heterocyclic modulators of lipid synthesis and combinations thereof
US9994550B2 (en) 2014-01-07 2018-06-12 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis for use against cancer and viral infections
CN105980376B (en) * 2014-01-07 2019-03-22 3-V生物科学股份有限公司 Lipid synthesis heterocyclic modulators for being used for cancer and virus infection
KR102335142B1 (en) 2014-01-07 2021-12-06 새지메트 바이오사이언시스, 인코포레이티드 Heterocyclic modulators of lipid synthesis for use against cancer and viral infections
CN105980376A (en) * 2014-01-07 2016-09-28 3-V生物科学股份有限公司 Heterocyclic modulators of lipid synthesis for use against cancer and viral infections
WO2015105860A1 (en) * 2014-01-07 2015-07-16 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis for use against cancer and viral infections
KR20160105416A (en) * 2014-01-07 2016-09-06 3-브이 바이오사이언시스, 인코포레이티드 Heterocyclic modulators of lipid synthesis for use against cancer and viral infections
RU2766087C2 (en) * 2014-01-07 2022-02-07 Сагимет Байосайенсиз Инк. Heterocyclic modulators of lipid synthesis for use against cancer and viral infections
JP2017502083A (en) * 2014-01-07 2017-01-19 3−ブイ・バイオサイエンシーズ・インコーポレイテッド3−V Biosciences,Inc. Heterocyclic modulators of lipid synthesis for use against cancer and viral infections
US10363249B2 (en) 2014-08-15 2019-07-30 3-V Biosciences, Inc. Fatty acid synthase inhibitor for use in the treatment of drug resistant cancer
RU2774758C2 (en) * 2015-03-19 2022-06-22 Сагимет Байосайенсиз Инк. Heterocyclic lipid synthesis modulators
US10189822B2 (en) 2015-03-19 2019-01-29 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
US11034690B2 (en) 2016-11-11 2021-06-15 Saginiet Biosciences Inc. Heterocyclic modulators of lipid synthesis
US10875848B2 (en) 2018-10-10 2020-12-29 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US11299484B2 (en) 2018-10-10 2022-04-12 Forma Therapeutics, Inc. Inhibiting fatty acid synthase (FASN)
US11267805B2 (en) 2018-10-29 2022-03-08 Forma Therapeutics, Inc. Solid forms of (4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl) piperazine-1-yl)(1-hydroxycyclopropyl)methanone
US10793554B2 (en) 2018-10-29 2020-10-06 Forma Therapeutics, Inc. Solid forms of 4-(2-fluoro-4-(1-methyl-1H-benzo[d]imidazol-5-yl)benzoyl)piperazin-1-yl)(1-hydroxycyclopropyl)methanone
WO2021130723A1 (en) * 2019-12-24 2021-07-01 Cadila Healthcare Limited Novel compounds suitable for the treatment of dyslipidemia

Similar Documents

Publication Publication Date Title
WO2008075077A1 (en) Piperidine derivatives for the treatment of obesity
US20090118332A1 (en) Therapeutic Agents - 551
US20090105305A1 (en) Therapeutic Agents - 550
JP7093438B2 (en) ERK1 and ERK2 heterocyclic inhibitors and their use in cancer treatment
WO2008059214A1 (en) Bisamlde derivatives and use thereof as fatty acid synthase inhibitors
EP3626713B1 (en) Cyclopropylamines for use as lsd1 inhibitors
AU2005223424B2 (en) Aryl and heteroaryl-piperidinecarboxylate derivatives, the preparation and the use thereof in the form of FAAH enzyme inhibitors
EP3105219B1 (en) Cyclopropylamines as lsd1 inhibitors
EP1828172B1 (en) Pyrrole derivatives having crth2 receptor antagonist activity
EP1018879B1 (en) Nitrosated and nitrosylated alpha-adrenergic receptor antagonist compounds, compositions and their uses
AU722480B2 (en) Nitrosated and nitrosylated phosphodiesterase inhibitor compounds, compositions and their uses
KR102232742B1 (en) Indole carboxamide derivatives as p2x7 receptor antagonists
CA2718123A1 (en) Quinazoline derivatives as raf kinase modulators and methods of use thereof
KR20090007371A (en) Piperidinyl substituted pyrrolidinones as inhibitors of 11-beta-hydroxysteroid dehydrogenase 1
SG176042A1 (en) 7-aza-spiro[3.5]nonane-7-carboxylate derivatives, preparation thereof, and therapeutic use thereof
EP1940823A2 (en) Substituted 1-amino-phthalzine derivatives, preparation and therapeutic use thereof
BRPI0707869A2 (en) azacyclyl substituted arylthienopyrimidinones, process for their preparation and their use as medicines
KR20110034020A (en) Alkyl thiazole carbamate derivatives, preparation thereof, and use thereof as faah enzyme inhibitors
WO2013170072A2 (en) Compounds for the treatment of neurological disorders
JP4837800B2 (en) Quinuclidine derivatives as muscarinic M3 receptor antagonists
EP1657241A1 (en) Novel anthranilamide pyridinureas as VEGF receptor kinase inhibitors
EP3436458B1 (en) Fused pentacyclic imidazole derivatives as modulators of tnf activity
KR20220053622A (en) Alpha-D-galactopyranoside derivatives
CN103906734A (en) Substituted methanesulfonamide derivatives as vanilloid receptor ligands
CN109384785B (en) Pyrrolopyridinone derivatives, preparation method and medical application thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07848661

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07848661

Country of ref document: EP

Kind code of ref document: A1