WO2008081208A1 - Piperidine gpcr agonists - Google Patents

Piperidine gpcr agonists Download PDF

Info

Publication number
WO2008081208A1
WO2008081208A1 PCT/GB2008/050014 GB2008050014W WO2008081208A1 WO 2008081208 A1 WO2008081208 A1 WO 2008081208A1 GB 2008050014 W GB2008050014 W GB 2008050014W WO 2008081208 A1 WO2008081208 A1 WO 2008081208A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutically acceptable
acceptable salt
compound according
alkyl
formula
Prior art date
Application number
PCT/GB2008/050014
Other languages
French (fr)
Inventor
Matthew Colin Thor Fyfe
Revathy Perpetua Jeevaratnam
John Keily
Simon Andrew Swain
Original Assignee
Prosidion 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
Priority claimed from GB0700126A external-priority patent/GB0700126D0/en
Priority claimed from GB0708748A external-priority patent/GB0708748D0/en
Priority claimed from GB0719765A external-priority patent/GB0719765D0/en
Application filed by Prosidion Limited filed Critical Prosidion Limited
Priority to BRPI0806312-5A priority Critical patent/BRPI0806312A2/en
Priority to JP2009544451A priority patent/JP2010514832A/en
Priority to EA200900878A priority patent/EA016507B1/en
Priority to US12/522,030 priority patent/US20100048625A1/en
Priority to EP08702097A priority patent/EP2114936A1/en
Priority to CA002674360A priority patent/CA2674360A1/en
Publication of WO2008081208A1 publication Critical patent/WO2008081208A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/12Ophthalmic agents for cataracts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Definitions

  • the present invention is directed to G-protein coupled receptor (GPCR) agonists.
  • GPCR G-protein coupled receptor
  • the present invention is directed to GPCR agonists that are useful for the treatment of obesity, e.g. as regulators of satiety, metabolic syndrome and for the treatment of diabetes.
  • Obesity is characterized by an excessive adipose tissue mass relative to body size.
  • body fat mass is estimated by the body mass index (BMI; weight(kg)/height(m) 2 ), or waist circumference.
  • BMI body mass index
  • Individuals are considered obese when the BMI is greater than 30 and there are established medical consequences of being overweight. It has been an accepted medical view for some time that an increased body weight, especially as a result of abdominal body fat, is associated with an increased risk for diabetes, hypertension, heart disease, and numerous other health complications, such as arthritis, stroke, gallbladder disease, muscular and respiratory problems, back pain and even certain cancers.
  • Drugs aimed at the pathophysiology associated with insulin dependent Type I diabetes and non-insulin dependent Type II diabetes have many potential side effects and do not adequately address the dyslipidaemia and hyperglycaemia in a high proportion of patients. Treatment is often focused at individual patient needs using diet, exercise, hypoglycaemic agents and insulin, but there is a continuing need for novel antidiabetic agents, particularly ones that may be better tolerated with fewer adverse effects.
  • metabolic syndrome places people at high risk of coronary artery disease, and is characterized by a cluster of risk factors including central obesity (excessive fat tissue in the abdominal region), glucose intolerance, high triglycerides and low HDL cholesterol, and high blood pressure.
  • central obesity excessive fat tissue in the abdominal region
  • glucose intolerance high triglycerides
  • low HDL cholesterol high blood pressure
  • Myocardial ischemia and microvascular disease is an established morbidity associated with untreated or poorly controlled metabolic syndrome.
  • GPRl 19 (previously referred to as GPRl 16) is a GPCR identified as SNORF25 in WO00/50562 which discloses both the human and rat receptors, US 6,468,756 also discloses the mouse receptor (accession numbers: AAN95194 (human), AAN95195 (rat) and ANN95196 (mouse)).
  • GPRl 19 is expressed in the pancreas, small intestine, colon and adipose tissue.
  • the expression profile of the human GPRl 19 receptor indicates its potential utility as a target for the treatment of obesity and diabetes.
  • International patent applications WO2005/061489, WO2006/070208, WO2006/067531 and WO2006/067532 disclose heterocyclic derivatives as GPRl 19 receptor agonists.
  • International patent applications PCT/GB2006/050176, PCT/GB2006/050177, PCT/GB2006/050178 and PCT/GB2006/050182 disclose further GPRl 19 receptor agonists.
  • the present invention relates to agonists of GPRl 19 which are useful for the treatment of obesity e.g. as peripheral regulators of satiety, metabolic syndrome and for the treatment of diabetes.
  • (I) or pharmaceutically acceptable salts thereof are agonists of GPRl 19 and are useful for the prophylactic or therapeutic treatment of obesity and diabetes.
  • the present invention is directed to a compound of formula (I), or a pharmaceutically acceptable salt thereof:
  • R 1 is SO 2 R 5 , NR 6 R 7 , or CONR 6 R 7 ;
  • R is hydrogen or methyl
  • R 3 is hydrogen or methyl
  • R 4 is C 2 -S alkyl
  • R 5 is Ci_ 3 alkyl
  • R 6 and R 7 are independently hydrogen, C M alkyl, which may optionally be substituted by halo, hydroxy, Ci_ 4 alkoxy-, aryloxy-, arylCi_ 4 alkoxy-, Ci_ 4 alkylS(O) m -, C 3 . 7 heterocyclyl, N(R 8 ) 2 or -C(O)OR 9 ; or may be C 3 . 7 cycloalkyl, aryl, heterocyclyl or heteroaryl, wherein the cyclic groups may be substituted with one or more substituents selected from halo, C 1 .
  • R 6 and R 7 may form a 5- or 6-membered heterocyclic ring optionally substituted by hydroxy, C 1 . 4 alkyl or C 1 . 4 hydroxyalkyl and optionally containing a further heteroatom selected from O and NR 8 ;
  • R 8 are independently hydrogen or C 1 . 4 alkyl; or a group N(R 8 ) 2 may form a 4- to 7- membered heterocyclic ring optionally containing a further heteroatom selected from O and NR 8 ;
  • R 9 is hydrogen or C 1 . 4 alkyl; and m is 0, 1 or 2.
  • X is O and in another Y is O.
  • X is preferably O.
  • Y is preferably N.
  • R 2 is preferably methyl.
  • R 3 is hydrogen and in another R 3 is methyl.
  • R 3 is methyl
  • the stereocentre created preferably has the (R)-configuration.
  • R 4 is preferably C3. 4 alkyl, particularly w-propyl, isopropyl, or tert-butyl, even more preferably C3 alkyl, especially isopropyl.
  • R 5 is preferably methyl.
  • R 6 is preferably hydrogen, Ci_3alkyl, or C 2 -3alkyl substituted by hydroxy and R 7 is preferably hydrogen.
  • R 6 is C 2 -3alkyl substituted by hydroxy, it may be substituted by one or more, e.g. 1 or 2, preferably 1, hydroxy group.
  • R 6 is more preferably Ci_3alkyl or C 2 -3alkyl substituted by hydroxy, more preferably C 2 3 alkyl substituted by hydroxy, e.g. 2-hydroxyethyl, 2-hydroxy-l-methylethyl, 2,3- dihydroxypropyl or 2-hydroxy-l-hydroxymethylethyl, preferably 2-hydroxyethyl or 2-hydroxy- l-methylethyl, even more preferably 2-hydroxy-l-methylethyl, especially (R)-2-hydroxy-l- methylethyl.
  • preferred compounds of this invention include those in which several or each variable in formula (I) is selected from the preferred, more preferred or particularly listed groups for each variable. Therefore, this invention is intended to include all combinations of preferred, more preferred and particularly listed groups.
  • alkyl means carbon chains which may be linear or branched or combinations thereof. Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec- and tert-butyl and pentyl.
  • halo includes fluorine, chlorine, bromine, and iodine atoms, in particular fluorine or chlorine, especially fluorine.
  • Compounds described herein may contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers.
  • the present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof.
  • the above formula (I) is shown without a definitive stereochemistry at certain positions.
  • the present invention includes all stereoisomers of formula (I) and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
  • the present invention includes any possible solvates and polymorphic forms.
  • a type of a solvent that forms the solvate is not particularly limited so long as the solvent is pharmacologically acceptable.
  • water, ethanol, propanol, acetone or the like can be used.
  • pharmaceutically acceptable salts refers to salts prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • Such acids include, for example, hydrochloric, methanesulfonic, sulfuric, p-toluenesulfonic acid and the like.
  • the compounds of formula (I) are intended for pharmaceutical use they are preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure, especially at least 98% pure (% are on a weight for weight basis).
  • the compounds of formula (I) can be prepared as described below.
  • PG represents a protecting group
  • G is a substituted oxadiazole as defined above
  • R 1 , R 2 , R 3 and R 4 are also as defined above.
  • a suitable catalyst is [Rh(norbornadiene) 2 ]BF 4 and (5)-l-[(R)-2-(di-tert-butylphosphino)ferrocenyl]- ethylbis(2-methylphenyl)phosphine.
  • Compounds of formula (IV) can then be obtained by reduction of the carboxylic acids of formula (III) under standard conditions, for example borane in a suitable solvent such as THF. Removal of the protecting group is then achieved under conditions well known to those with skill in the art.
  • Compounds of formula (VII) can be prepared from compounds of formula (V) under standard conditions. For example, treatment of compounds of formula (V) with cyanogen bromide followed by condensation of the resultant cyanamide (VI) with a compound of formula (IX) under standard conditions yields compounds of formula (VII) where X is O.
  • Compounds of formula (IX) are either commercially available, or readily prepared from the corresponding carboxylic acids using well known techniques.
  • synthesis of the regioisomeric oxadiazole, where Y is O can be achieved by heating compounds of formula (VI) with hydroxylamine to give N-hydroxyguanidines of formula (VIII) that may be condensed with a carboxylic acid of formula (X) under suitable conditions. Acids of formula (X) are commercially available.
  • the compounds of formula (I) may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000, compounds and more preferably 10 to 100 compounds of formula (I).
  • Compound libraries may be prepared by a combinatorial "split and mix” approach or by multiple parallel synthesis using either solution or solid phase chemistry, using procedures known to those skilled in the art.
  • labile functional groups in the intermediate compounds e.g. hydroxy, carboxy and amino groups
  • the protecting groups may be removed at any stage in the synthesis of the compounds of formula (I) or may be present on the final compound of formula (I).
  • a comprehensive discussion of the ways in which various labile functional groups may be protected and methods for cleaving the resulting protected derivatives is given in, for example, Protective Groups in Organic Chemistry, T. W. Greene and P.G.M. Wuts, (1991) Wiley-Interscience, New York, 2 nd edition.
  • the compounds of formula (I) may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000, compounds and more preferably 10 to 100 compounds of formula (I).
  • Compound libraries may be prepared by a combinatorial "split and mix” approach or by multiple parallel synthesis using either solution or solid phase chemistry, using procedures known to those skilled in the art.
  • labile functional groups in the intermediate compounds e.g. hydroxy, carboxy and amino groups
  • the protecting groups may be removed at any stage in the synthesis of the compounds of formula (I) or may be present on the final compound of formula (I).
  • a comprehensive discussion of the ways in which various labile functional groups may be protected and methods for cleaving the resulting protected derivatives is given in, for example, Protective Groups in Organic Chemistry, T.W. Greene and P.G.M. Wuts, (1991) Wiley-Interscience, New York, 2 nd edition.
  • the compounds of formula (I) are useful as GPRl 19 agonists, e.g. for the treatment and/or prophylaxis of obesity and diabetes.
  • the compounds of formula (I) will generally be administered in the form of a pharmaceutical composition.
  • the invention also provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, for use as a pharmaceutical.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), in combination with a pharmaceutically acceptable carrier.
  • the composition is comprised of a pharmaceutically acceptable carrier and a non-toxic therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • the invention also provides a pharmaceutical composition for the treatment of disease by modulating GPRl 19, resulting in the prophylactic or therapeutic treatment of obesity, e.g. by regulating satiety, or for the treatment of diabetes, comprising a pharmaceutically acceptable carrier and a non-toxic therapeutically effective amount of compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • compositions may optionally comprise other therapeutic ingredients or adjuvants.
  • the compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered.
  • the pharmaceutical compositions may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
  • the compounds of formula (I), or pharmaceutically acceptable salts thereof can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g. oral or parenteral (including intravenous).
  • compositions can be presented as discrete units suitable for oral administration such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient. Further, the compositions can be presented as a powder, as granules, as a solution, as a suspension in an aqueous liquid, as a non-aqueous liquid, as an oil-in- water emulsion, or as a water-in-oil liquid emulsion.
  • the compound of formula (I), or a pharmaceutically acceptable salt thereof may also be administered by controlled release means and/or delivery devices.
  • the compositions may be prepared by any of the methods of pharmacy.
  • such methods include a step of bringing into association the active ingredient with the carrier that constitutes one or more necessary ingredients.
  • the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both. The product can then be conveniently shaped into the desired presentation.
  • the compounds of formula (I), or pharmaceutically acceptable salts thereof, can also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds.
  • the pharmaceutical carrier employed can be, for example, a solid, liquid, or gas.
  • solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid.
  • liquid carriers are sugar syrup, peanut oil, olive oil, and water.
  • gaseous carriers include carbon dioxide and nitrogen.
  • any convenient pharmaceutical media may be employed.
  • water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like may be used to form oral liquid preparations such as suspensions, elixirs and solutions; while carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be used to form oral solid preparations such as powders, capsules and tablets. Because of their ease of administration, tablets and capsules are the preferred oral dosage units whereby solid pharmaceutical carriers are employed.
  • tablets may be coated by standard aqueous or nonaqueous techniques.
  • a tablet containing the composition of this invention may be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants.
  • Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free- flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
  • Each tablet preferably contains from about 0.05mg to about 5g of the active ingredient and each cachet or capsule preferably containing from about 0.05mg to about 5g of the active ingredient.
  • a formulation intended for the oral administration to humans may contain from about 0.5mg to about 5g of active agent, compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95 percent of the total composition.
  • Unit dosage forms will generally contain between from about lmg to about 2g of the active ingredient, typically 25mg, 50mg, lOOmg, 200mg, 300mg, 400mg, 500mg, 600mg, 800mg, or lOOOmg.
  • compositions of the present invention suitable for parenteral administration may be prepared as solutions or suspensions of the active compounds in water.
  • a suitable surfactant can be included such as, for example, hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms.
  • compositions of the present invention suitable for injectable use include sterile aqueous solutions or dispersions.
  • the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions.
  • the final injectable form must be sterile and must be effectively fluid for easy syringability.
  • the pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g. glycerol, propylene glycol and liquid polyethylene glycol), vegetable oils, and suitable mixtures thereof.
  • compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, or the like. Further, the compositions can be in a form suitable for use in transdermal devices. These formulations may be prepared, using a compound of formula (I), or a pharmaceutically acceptable salt thereof, via conventional processing methods. As an example, a cream or ointment is prepared by admixing hydrophilic material and water, together with about 5wt% to about 10wt% of the compound, to produce a cream or ointment having a desired consistency.
  • compositions of this invention can be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories may be conveniently formed by first admixing the composition with the softened or melted carrier(s) followed by chilling and shaping in molds.
  • the pharmaceutical formulations described above may include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti- oxidants) and the like.
  • additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti- oxidants) and the like.
  • other adjuvants can be included to render the formulation isotonic with the blood of the intended recipient.
  • dosage levels on the order of O.Olmg/kg to about 150mg/kg of body weight per day are useful in the treatment of the above- indicated conditions, or alternatively about 0.5mg to about 7g per patient per day.
  • obesity may be effectively treated by the administration of from about 0.01 to 50mg of the compound per kilogram of body weight per day, or alternatively about 0.5mg to about 3.5g per patient per day.
  • the compounds of formula (I) may be used in the treatment of diseases or conditions in which GPRl 19 plays a role.
  • the invention also provides a method for the treatment of a disease or condition in which GPRl 19 plays a role comprising a step of administering to a subject in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • Diseases or conditions in which GPRl 19 plays a role include obesity and diabetes.
  • the treatment of obesity is intended to encompass the treatment of diseases or conditions such as obesity and other eating disorders associated with excessive food intake e.g. by reduction of appetite and body weight, maintenance of weight reduction and prevention of rebound and diabetes (including Type 1 and Type 2 diabetes, impaired glucose tolerance, insulin resistance and diabetic complications such as neuropathy, nephropathy, retinopathy, cataracts, cardiovascular complications and dyslipidaemia).
  • the compounds of the invention may also be used for treating metabolic diseases such as metabolic syndrome (syndrome X), impaired glucose tolerance, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low HDL levels and hypertension.
  • the compounds of the invention may offer advantages over compounds acting via different mechanisms for the treatment of the above mentioned disorders in that they may offer beta-cell protection, increased cAMP and insulin secretion and also slow gastric emptying.
  • the compounds of the invention may also be used for treating conditions characterised by low bone mass such asosteopenia, osteoporosis, rheumatoid arthritis, osteoarthritis, periodontal disease, alveolar bone loss, osteotomy bone loss, childhood idiopathic bone loss, Paget's disease, bone loss due to metastatic cancer, osteolytic lesions, curvature of the spine and loss of height.
  • low bone mass such asosteopenia, osteoporosis, rheumatoid arthritis, osteoarthritis, periodontal disease, alveolar bone loss, osteotomy bone loss, childhood idiopathic bone loss, Paget's disease, bone loss due to metastatic cancer, osteolytic lesions, curvature of the spine and loss of height.
  • the invention also provides a method for the regulation of satiety comprising a step of administering to a subject in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • the invention also provides a method for the treatment of obesity comprising a step of administering to a subject in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • the invention also provides a method for the treatment of diabetes, including Type 1 and Type 2 diabetes, particularly type 2 diabetes, comprising a step of administering to a patient in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • the invention also provides a method for the treatment of metabolic syndrome (syndrome X), impaired glucose tolerance, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low HDL levels or hypertension comprising a step of administering to a patient in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • metabolic syndrome sekunder X
  • impaired glucose tolerance hyperlipidemia
  • hypertriglyceridemia hypercholesterolemia
  • low HDL levels or hypertension comprising a step of administering to a patient in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • the invention also provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of a condition as defined above.
  • the invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a condition as defined above.
  • treatment includes both therapeutic and prophylactic treatment.
  • the compounds of formula (I) may exhibit advantageous properties compared to known GPR 119 agonists, for example, the compounds may exhibit improved potency or stability, or improved solubility thus improving absorption properties and bioavailability, or other advantageous properties for compounds to be used as pharmaceuticals.
  • the compounds of formula (I), or pharmaceutically acceptable salts thereof, may be administered alone or in combination with one or more other therapeutically active compounds.
  • the other therapeutically active compounds may be for the treatment of the same disease or condition as the compounds of formula (I) or a different disease or condition.
  • the therapeutically active compounds may be administered simultaneously, sequentially or separately.
  • the compounds of formula (I) may be administered with other active compounds for the treatment of obesity and/or diabetes, for example insulin and insulin analogs, gastric lipase inhibitors, pancreatic lipase inhibitors, sulfonyl ureas and analogs, biguanides, ⁇ 2 agonists, glitazones, PPAR- ⁇ agonists, mixed PPAR- ⁇ / ⁇ agonists, RXR agonists, fatty acid oxidation inhibitors, ⁇ -glucosidase inhibitors, dipeptidyl peptidase IV inhibitors, GLP-I agonists e.g.
  • active compounds for the treatment of obesity and/or diabetes for example insulin and insulin analogs, gastric lipase inhibitors, pancreatic lipase inhibitors, sulfonyl ureas and analogs, biguanides, ⁇ 2 agonists, glitazones, PPAR- ⁇ agonists, mixed PPAR- ⁇ / ⁇ agonists,
  • GLP-I analogues and mimetics ⁇ -agonists, phosphodiesterase inhibitors, lipid lowering agents, glycogen phosphorylase inhibitors, antiobesity agents e.g. pancreatic lipase inhibitors, MCH-I antagonists and CB-I antagonists (or inverse agonists), amylin antagonists, lipoxygenase inhibitors, somostatin analogs, glucokinase activators, glucagon antagonists, insulin signalling agonists, PTPlB inhibitors, gluconeogenesis inhibitors, antilypolitic agents, GSK inhibitors, galanin receptor agonists, anorectic agents, CCK receptor agonists, leptin, serotonergic/dopaminergic antiobesity drugs, reuptake inhibitors e.g.
  • Combination therapy comprising the administration of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and at least one other antiobesity agent represents a further aspect of the invention.
  • the present invention also provides a method for the treatment of obesity in a mammal, such as a human, which method comprises administering an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and another antiobesity agent, to a mammal in need thereof.
  • the invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and another antiobesity agent for the treatment of obesity.
  • the invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in combination with another antiobesity agent, for the treatment of obesity.
  • the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the other antiobesity agent(s) may be co-administered or administered sequentially or separately.
  • Co-administration includes administration of a formulation which includes both the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the other antiobesity agent(s), or the simultaneous or separate administration of different formulations of each agent. Where the pharmacological profiles of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the other antiobesity agent(s) allow it, coadministration of the two agents may be preferred.
  • the invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and another antiobesity agent in the manufacture of a medicament for the treatment of obesity.
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and another antiobesity agent, and a pharmaceutically acceptable carrier.
  • the invention also encompasses the use of such compositions in the methods described above.
  • GPRl 19 agonists are of particular use in combination with centrally acting antiobesity agents.
  • the other antiobesity agent for use in the combination therapies according to this aspect of the invention is preferably a CB-I modulator, e.g. a CB-I antagonist or inverse agonist.
  • CB-I modulators include SR141716 (rimonabant) and SLV-319 ((45)-(-)-3-(4- chlorophenyl)-N-methyl-N-[(4-chlorophenyl)sulfonyl]-4-phenyl-4,5-dihydro-lH-pyrazole-l- carboxamide); as well as those compounds disclosed in EP576357, EP656354, WO 03/018060, WO 03/020217, WO 03/020314, WO 03/026647, WO 03/026648, WO 03/027076, WO 03/040105, WO 03/051850, WO 03/051851, WO 03/053431, WO 03/063781, WO 03/
  • GPRl 19 has been suggested to play a role
  • diseases or conditions in which GPRl 19 has been suggested to play a role include those described in WO 00/50562 and US 6,468,756, for example cardiovascular disorders, hypertension, respiratory disorders, gestational abnormalities, gastrointestinal disorders, immune disorders, musculoskeletal disorders, depression, phobias, anxiety, mood disorders and Alzheimer's disease.
  • Example 1 5- ⁇ 3-[l-(3 -Isopropyl- [ 1 ,2,4]oxadiazol-5 -yl)piperidin-4-yl]propoxy ⁇ -3 -methyl- pyridine-2-carboxylic acid ((R)-2-hydroxy-l-methylethyl)amide
  • the biological activity of the compounds of the invention may be tested in the following assay systems: Yeast Reporter Assay
  • yeast cell-based reporter assays have previously been described in the literature (e.g. see Miret J. J. et al, 2002, J. Biol. Chem., 277:6881-6887; Campbell R.M. et al, 1999, Bioorg. Med. Chem. Lett, 9:2413-2418; King K. et al, 1990, Science, 250:121-123); WO 99/14344; WO 00/12704; and US 6,100,042). Briefly, yeast cells have been engineered such that the endogenous yeast G-alpha (GPAl) has been deleted and replaced with G-protein chimeras constructed using multiple techniques.
  • G-alpha G-alpha
  • yeast GPCR Ste3 has been deleted to allow for heterologous expression of a mammalian GPCR of choice.
  • elements of the pheromone signaling transduction pathway which are conserved in eukaryotic cells (for example, the mitogen-activated protein kinase pathway), drive the expression of Fusl.
  • ⁇ -galactosidase LacZ
  • Fuslp Fusl promoter
  • Yeast cells were transformed by an adaptation of the lithium acetate method described by Agatep et al, (Agatep, R. et al, 1998, Transformation of Saccharomyces cerevisiae by the lithium acetate/single-stranded carrier DNA/polyethylene glycol (LiAc/ss-DNA/PEG) protocol. Technical Tips Online, Trends Journals, Elsevier). Briefly, yeast cells were grown overnight on yeast tryptone plates (YT).
  • Carrier single-stranded DNA (lO ⁇ g), 2 ⁇ g of each of two Fuslp- LacZ reporter plasmids (one with URA selection marker and one with TRP), 2 ⁇ g of GPRl 19 (human or mouse receptor) in yeast expression vector (2 ⁇ g origin of replication) and a lithium acetate/ polyethylene glycol/ TE buffer was pipetted into an Eppendorf tube.
  • the yeast expression plasmid containing the receptor/ no receptor control has a LEU marker.
  • Yeast cells were inoculated into this mixture and the reaction proceeds at 30 0 C for 60min.
  • the yeast cells were then heat-shocked at 42°C for 15min.
  • the cells were then washed and spread on selection plates.
  • the selection plates are synthetic defined yeast media minus LEU, URA and TRP (SD- LUT). After incubating at 30 0 C for 2-3 days, colonies that grow on the selection plates were then tested in the LacZ assay.
  • yeast cells carrying the human or mouse GPRl 19 receptor were grown overnight in liquid SD-LUT medium to an unsaturated concentration (i.e. the cells were still dividing and had not yet reached stationary phase). They were diluted in fresh medium to an optimal assay concentration and 90 ⁇ l of yeast cells added to 96-well black polystyrene plates (Costar). Compounds, dissolved in DMSO and diluted in a 10% DMSO solution to 1OX concentration, were added to the plates and the plates placed at 30 0 C for 4h. After 4h, the substrate for the ⁇ -galactosidase was added to each well.
  • Fluorescein di ⁇ -D-galactopyranoside
  • FDG Fluorescein di
  • a substrate for the enzyme that releases fluorescein allowing a fluorimetric read-out.
  • 20 ⁇ l per well of 500 ⁇ M FDG/2.5% Triton XlOO was added (the detergent was necessary to render the cells permeable).
  • 20 ⁇ l per well of IM sodium carbonate was added to terminate the reaction and enhance the fluorescent signal.
  • the plates were then read in a fluorimeter at 485/535nm.
  • the compounds of the invention give an increase in fluorescent signal of at least ⁇ 1.5- fold that of the background signal (i.e. the signal obtained in the presence of 1% DMSO without compound).
  • Compounds of the invention which give an increase of at least 5-fold may be preferred.
  • a stable cell line expressing recombinant human GPRl 19 was established and this cell line may be used to investigate the effect of compounds of the invention on intracellular levels of cyclic AMP (cAMP).
  • cAMP cyclic AMP
  • the cell monolayers are washed with phosphate buffered saline and stimulated at 37°C for 30min with various concentrations of compound in stimulation buffer plus 1% DMSO. Cells are then lysed and cAMP content determined using the Perkin Elmer AlphaScreenTM (Amplified Luminescent Proximity Homogeneous Assay) cAMP kit. Buffers and assay conditions are as described in the manufacturer's protocol.
  • Test compounds and reference compounds are dosed by appropriate routes of administration (e.g. intraperitoneally or orally) and measurements made over the following 24 h.
  • Rats are individually housed in polypropylene cages with metal grid floors at a temperature of 21 ⁇ 4°C and 55 ⁇ 20% humidity. Polypropylene trays with cage pads are placed beneath each cage to detect any food spillage. Animals are maintained on a reverse phase light-dark cycle (lights off for 8 h from 09.30-17.30 h) during which time the room is illuminated by red light.
  • Animals have free access to a standard powdered rat diet and tap water during a two week acclimatization period.
  • the diet is contained in glass feeding jars with aluminum lids. Each lid has a 3-4 cm hole in it to allow access to the food.
  • Animals, feeding jars and water bottles are weighed (to the nearest 0.1 g) at the onset of the dark period. The feeding jars and water bottles are subsequently measured 1, 2, 4, 6 and 24 h after animals are dosed with a compound of the invention and any significant differences between the treatment groups at baseline compared to vehicle-treated controls.
  • HIT-Tl 5 cells (passage 60) were obtained from ATCC, and were cultured in RPMI 1640 medium supplemented with 10% fetal calf serum and 3OnM sodium selenite. All experiments were done with cells at less than passage 70, in accordance with the literature, which describes altered properties of this cell line at passage numbers above 81 (Zhang HJ, Walseth TF, Robertson RP. Insulin secretion and cAMP metabolism in HIT cells. Reciprocal and serial passage-dependent relationships. Diabetes. 1989 Jan;38(l):44-8).
  • HIT-T15 cells were plated in standard culture medium in 96-well plates at 100,000 cells/ 0.1ml/ well and cultured for 24 hr and the medium was then discarded. Cells were incubated for 15min at room temperature with lOO ⁇ l stimulation buffer (Hanks buffered salt solution, 5mM HEPES, 0.5mM IBMX, 0.1% BSA, pH 7.4). This was discarded and replaced with compound dilutions over the range 0.001, 0.003, 0.01, 0.03, 0.1, 0.3, 1, 3, 10, 30 ⁇ M in stimulation buffer in the presence of 0.5% DMSO. Cells were incubated at room temperature for 30min.
  • lOO ⁇ l stimulation buffer Hors buffered salt solution, 5mM HEPES, 0.5mM IBMX, 0.1% BSA, pH 7.4
  • 75ul lysis buffer (5mM HEPES, 0.3% Tween-20, 0.1% BSA, pH 7.4) was added per well and the plate was shaken at 900 rpm for 20 min. Particulate matter was removed by centrifugation at 3000rpm for 5min, then the samples were transferred in duplicate to 384-well plates, and processed following the Perkin Elmer AlphaScreen cAMP assay kit instructions. Briefly 25 ⁇ l reactions were set up containing 8 ⁇ l sample, 5 ⁇ l acceptor bead mix and 12 ⁇ l detection mix, such that the concentration of the final reaction components is the same as stated in the kit instructions. Reactions were incubated at room temperature for 150min, and the plate was read using a Packard Fusion instrument.
  • Measurements for cAMP were compared to a standard curve of known cAMP amounts (0.01, 0.03, 0.1, 0.3, 1, 3, 10, 30, 100, 300, 1000 nM) to convert the readings to absolute cAMP amounts. Data was analysed using XLfit 3 software.
  • Representative compounds of the invention were found to increase cAMP at an EC 5 O of less than 10 ⁇ M. Compounds showing an EC 5 O of less than 1 ⁇ M in the cAMP assay may be preferred.
  • HIT-Tl 5 cells are plated in standard culture medium in 12-well plates at 106 cells/ 1 ml/ well and cultured for 3 days and the medium is then discarded.
  • Cells are washed x 2 with supplemented Krebs-Ringer buffer (KRB) containing 119 mM NaCl, 4.74 mM KCl, 2.54 mM CaCl 2 , 1.19 mM MgSO 4 , 1.19 mM KH2PO4, 25 mM NaHCO 3 , 1OmM HEPES at pH 7.4 and 0.1% bovine serum albumin.
  • KRB Krebs-Ringer buffer
  • Blood samples were then taken from the cut tip of the tail 5, 15, 30, 60, 120, and 180 min after GIc administration. Blood glucose levels were measured just after collection using a commercially available glucose-meter (OneTouch® UltraTM from Lifescan). Representative compounds of the invention statistically reduced the GIc excursion at doses of ⁇ 10 mg kg "1 .
  • GIc oral glucose
  • Food is withdrawn 5 h before administration of GIc and remains withdrawn throughout the study. Mice have free access to water during the study. A cut is made to the animals' tails, then blood (20 ⁇ L) is removed for measurement of basal GIc levels 45 min before administration of the GIc load.
  • mice are weighed and dosed orally with test compound or vehicle (20% aqueous hydroxypropyl-yS-cyclodextrin or 25% aqueous Gelucire 44/14) 30 min before the removal of an additional blood sample (20 ⁇ L) and treatment with the GIc load (2-5 g kg "1 p.o.). Blood samples (20 ⁇ L) are then taken 25, 50, 80, 120, and 180 min after GIc administration. The 20 ⁇ L blood samples for measurement of GIc levels are taken from the cut tip of the tail into disposable micro-pipettes (Dade Diagnostics Inc., Puerto Rico) and the sample added to 480 ⁇ L of haemolysis reagent.
  • test compound or vehicle 20% aqueous hydroxypropyl-yS-cyclodextrin or 25% aqueous Gelucire 44/14)
  • Duplicate 20 ⁇ L aliquots of the diluted haemolysed blood are then added to 180 ⁇ L of Trinders glucose reagent (Sigma enzymatic (Trinder) colorimetric method) in a 96-well assay plate. After mixing, the samples are left at rt for 30 min before being read against GIc standards (Sigma glucose/urea nitrogen combined standard set).

Abstract

Compounds of formula (I): or pharmaceutically acceptable salts thereof, are GPCR agonists and are useful as for the treatment of obesity and diabetes.

Description

PIPERIDINE GPCR AGONISTS
BACKGROUND OF THE INVENTION
The present invention is directed to G-protein coupled receptor (GPCR) agonists. In particular, the present invention is directed to GPCR agonists that are useful for the treatment of obesity, e.g. as regulators of satiety, metabolic syndrome and for the treatment of diabetes.
Obesity is characterized by an excessive adipose tissue mass relative to body size. Clinically, body fat mass is estimated by the body mass index (BMI; weight(kg)/height(m)2), or waist circumference. Individuals are considered obese when the BMI is greater than 30 and there are established medical consequences of being overweight. It has been an accepted medical view for some time that an increased body weight, especially as a result of abdominal body fat, is associated with an increased risk for diabetes, hypertension, heart disease, and numerous other health complications, such as arthritis, stroke, gallbladder disease, muscular and respiratory problems, back pain and even certain cancers.
Pharmacological approaches to the treatment of obesity have been mainly concerned with reducing fat mass by altering the balance between energy intake and expenditure. Many studies have clearly established the link between adiposity and the brain circuitry involved in the regulation of energy homeostasis. Direct and indirect evidence suggest that serotonergic, dopaminergic, adrenergic, cholinergic, endocannabinoid, opioid, and histaminergic pathways in addition to many neuropeptide pathways (e.g. neuropeptide Y and melanocortins) are implicated in the central control of energy intake and expenditure. Hypothalamic centres are also able to sense peripheral hormones involved in the maintenance of body weight and degree of adiposity, such as insulin and leptin, and fat tissue derived peptides.
Drugs aimed at the pathophysiology associated with insulin dependent Type I diabetes and non-insulin dependent Type II diabetes have many potential side effects and do not adequately address the dyslipidaemia and hyperglycaemia in a high proportion of patients. Treatment is often focused at individual patient needs using diet, exercise, hypoglycaemic agents and insulin, but there is a continuing need for novel antidiabetic agents, particularly ones that may be better tolerated with fewer adverse effects.
Similarly, metabolic syndrome (syndrome X) places people at high risk of coronary artery disease, and is characterized by a cluster of risk factors including central obesity (excessive fat tissue in the abdominal region), glucose intolerance, high triglycerides and low HDL cholesterol, and high blood pressure. Myocardial ischemia and microvascular disease is an established morbidity associated with untreated or poorly controlled metabolic syndrome.
There is a continuing need for novel antiobesity and antidiabetic agents, particularly ones that are well tolerated with few adverse effects.
GPRl 19 (previously referred to as GPRl 16) is a GPCR identified as SNORF25 in WO00/50562 which discloses both the human and rat receptors, US 6,468,756 also discloses the mouse receptor (accession numbers: AAN95194 (human), AAN95195 (rat) and ANN95196 (mouse)).
In humans, GPRl 19 is expressed in the pancreas, small intestine, colon and adipose tissue. The expression profile of the human GPRl 19 receptor indicates its potential utility as a target for the treatment of obesity and diabetes. International patent applications WO2005/061489, WO2006/070208, WO2006/067531 and WO2006/067532 disclose heterocyclic derivatives as GPRl 19 receptor agonists. International patent applications PCT/GB2006/050176, PCT/GB2006/050177, PCT/GB2006/050178 and PCT/GB2006/050182 (published after the priority date of the present application) disclose further GPRl 19 receptor agonists.
The present invention relates to agonists of GPRl 19 which are useful for the treatment of obesity e.g. as peripheral regulators of satiety, metabolic syndrome and for the treatment of diabetes.
SUMMARY OF THE INVENTION Compounds of formula (I):
Figure imgf000003_0001
(I) or pharmaceutically acceptable salts thereof, are agonists of GPRl 19 and are useful for the prophylactic or therapeutic treatment of obesity and diabetes.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to a compound of formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000003_0002
(I) wherein one of X and Y is O and the other is N;
R1 is SO2R5, NR6R7, or CONR6R7;
R is hydrogen or methyl;
R3 is hydrogen or methyl;
R4 is C2-S alkyl;
R5 is Ci_3alkyl;
R6 and R7 are independently hydrogen, CM alkyl, which may optionally be substituted by halo, hydroxy, Ci_4 alkoxy-, aryloxy-, arylCi_4 alkoxy-, Ci_4 alkylS(O)m-, C3.7 heterocyclyl, N(R8)2 or -C(O)OR9; or may be C3.7 cycloalkyl, aryl, heterocyclyl or heteroaryl, wherein the cyclic groups may be substituted with one or more substituents selected from halo, C 1.4 alkyl, Ci- 4 fluoroalkyl, OR6, CN, SO2CH3, N(R8)2 and NO2; or taken together R6 and R7 may form a 5- or 6-membered heterocyclic ring optionally substituted by hydroxy, C 1.4 alkyl or C 1.4 hydroxyalkyl and optionally containing a further heteroatom selected from O and NR8; R8 are independently hydrogen or C 1.4 alkyl; or a group N(R8)2 may form a 4- to 7- membered heterocyclic ring optionally containing a further heteroatom selected from O and NR8;
R9 is hydrogen or C 1.4 alkyl; and m is 0, 1 or 2.
In one embodiment of the invention X is O and in another Y is O.
X is preferably O.
Y is preferably N.
When R1 is CONHR6, R2 is preferably methyl.
In one embodiment of the invention R3 is hydrogen and in another R3 is methyl. When R3 is methyl, the stereocentre created preferably has the (R)-configuration.
R4 is preferably C3.4 alkyl, particularly w-propyl, isopropyl, or tert-butyl, even more preferably C3 alkyl, especially isopropyl.
R5 is preferably methyl.
R6 is preferably hydrogen, Ci_3alkyl, or C2-3alkyl substituted by hydroxy and R7 is preferably hydrogen.
When R6 is C2-3alkyl substituted by hydroxy, it may be substituted by one or more, e.g. 1 or 2, preferably 1, hydroxy group.
R6 is more preferably Ci_3alkyl or C2-3alkyl substituted by hydroxy, more preferably C2 3alkyl substituted by hydroxy, e.g. 2-hydroxyethyl, 2-hydroxy-l-methylethyl, 2,3- dihydroxypropyl or 2-hydroxy-l-hydroxymethylethyl, preferably 2-hydroxyethyl or 2-hydroxy- l-methylethyl, even more preferably 2-hydroxy-l-methylethyl, especially (R)-2-hydroxy-l- methylethyl.
While the preferred groups for each variable have generally been listed above separately for each variable, preferred compounds of this invention include those in which several or each variable in formula (I) is selected from the preferred, more preferred or particularly listed groups for each variable. Therefore, this invention is intended to include all combinations of preferred, more preferred and particularly listed groups.
Specific compounds of the invention which may be mentioned are those included in the Examples and pharmaceutically acceptable salts thereof.
As used herein, unless stated otherwise, "alkyl" means carbon chains which may be linear or branched or combinations thereof. Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec- and tert-butyl and pentyl.
The term "halo" includes fluorine, chlorine, bromine, and iodine atoms, in particular fluorine or chlorine, especially fluorine.
Compounds described herein may contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers. The present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. The above formula (I) is shown without a definitive stereochemistry at certain positions. The present invention includes all stereoisomers of formula (I) and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
When the compound of formula (I) and pharmaceutically acceptable salts thereof exist in the form of solvates or polymorphic forms, the present invention includes any possible solvates and polymorphic forms. A type of a solvent that forms the solvate is not particularly limited so long as the solvent is pharmacologically acceptable. For example, water, ethanol, propanol, acetone or the like can be used.
The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include, for example, hydrochloric, methanesulfonic, sulfuric, p-toluenesulfonic acid and the like.
Since the compounds of formula (I) are intended for pharmaceutical use they are preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure, especially at least 98% pure (% are on a weight for weight basis).
The compounds of formula (I) can be prepared as described below. PG represents a protecting group, G is a substituted oxadiazole as defined above, and R1, R2, R3 and R4 are also as defined above.
Compounds of formula (II), where PG is a suitable protecting group can be readily prepared from known compounds (Scheme 1). For example, the ethyl ester of compound (II) where PG is Boc has been previously reported (US Patent 6,518,423). Hydrogenation under standard conditions will yield the racemic compound of formula (III). Chiral reduction of the alkene under suitable conditions such as a hydrogenation in the presence of a chiral catalyst yields compounds of formula (III) in high enantiomeric excess. An example of a suitable catalyst is [Rh(norbornadiene)2]BF4 and (5)-l-[(R)-2-(di-tert-butylphosphino)ferrocenyl]- ethylbis(2-methylphenyl)phosphine. Compounds of formula (IV) can then be obtained by reduction of the carboxylic acids of formula (III) under standard conditions, for example borane in a suitable solvent such as THF. Removal of the protecting group is then achieved under conditions well known to those with skill in the art.
Scheme 1
Figure imgf000005_0001
II HI IV
The compound of formula (V) where R3= H is a known compound (Scheme 2, Siegel, M. G. et al. Tetrahedron 1999, 55, 11619-11639). Compounds of formula (VII) can be prepared from compounds of formula (V) under standard conditions. For example, treatment of compounds of formula (V) with cyanogen bromide followed by condensation of the resultant cyanamide (VI) with a compound of formula (IX) under standard conditions yields compounds of formula (VII) where X is O. Compounds of formula (IX) are either commercially available, or readily prepared from the corresponding carboxylic acids using well known techniques. Alternatively, synthesis of the regioisomeric oxadiazole, where Y is O, can be achieved by heating compounds of formula (VI) with hydroxylamine to give N-hydroxyguanidines of formula (VIII) that may be condensed with a carboxylic acid of formula (X) under suitable conditions. Acids of formula (X) are commercially available.
Scheme 2
Figure imgf000006_0001
Compounds of the formula (VII) may also be prepared by condensation of amine (V) with an oxadiazole chloride of formula (XI), as illustrated in Scheme 3 (Buscemi, S. et al. JCS Perkin I: Org. and Bioorg. Chem., 1988, 1313 and Adembri, G, et al. JCS Perkin I: Org. and Bioorg. Chem., 1981, 1703).
Scheme 3
Figure imgf000006_0002
V XI VII
Compounds of formula (I) where R1 is either a sulfone or amide can be formed as outlined in Scheme 4. The compound where R1 is methylsulfonyl and R2 is hydrogen has been reported previously (WO 2005/063738). Combining compounds of formula (VIII) and formula (VII) using Mitsunobu conditions, for example in a suitable solvent such as THF, at between O0C and room temperature, followed by the addition of triphenylphosphine and diisopropylazodicarboxylate yields compounds of formula (I). Compounds of formula (I) where R1 is an amide can be synthesized starting from the commercially available compound of formula (VIII) where R1 is cyano and R2 is methyl using chemistry well known to those with skill in the art. For example, Mitsunobu condensation of this compound with the alcohol of formula (VII) would give a cyanopyridine that could be transformed into the corresponding acid by hydrolysis, then onto the desired amides by standard amide bond-forming techniques. Aminopyridines of formula (I) may be prepared from the corresponding sulfones of formula (I) by displacement of alkanesulfmate by the appropriate amine on heating at an elevated temperature.
Scheme 4
Figure imgf000007_0001
Other compounds of formula (I) may be prepared by methods analogous to those described above or by methods known per se.
The compounds of formula (I) may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000, compounds and more preferably 10 to 100 compounds of formula (I). Compound libraries may be prepared by a combinatorial "split and mix" approach or by multiple parallel synthesis using either solution or solid phase chemistry, using procedures known to those skilled in the art.
During the synthesis of the compounds of formula (I), labile functional groups in the intermediate compounds, e.g. hydroxy, carboxy and amino groups, may be protected. The protecting groups may be removed at any stage in the synthesis of the compounds of formula (I) or may be present on the final compound of formula (I). A comprehensive discussion of the ways in which various labile functional groups may be protected and methods for cleaving the resulting protected derivatives is given in, for example, Protective Groups in Organic Chemistry, T. W. Greene and P.G.M. Wuts, (1991) Wiley-Interscience, New York, 2nd edition.
Other compounds of formula (I) may be prepared by methods analogous to those described above or by methods known per se.
The compounds of formula (I) may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000, compounds and more preferably 10 to 100 compounds of formula (I). Compound libraries may be prepared by a combinatorial "split and mix" approach or by multiple parallel synthesis using either solution or solid phase chemistry, using procedures known to those skilled in the art.
During the synthesis of the compounds of formula (I), labile functional groups in the intermediate compounds, e.g. hydroxy, carboxy and amino groups, may be protected. The protecting groups may be removed at any stage in the synthesis of the compounds of formula (I) or may be present on the final compound of formula (I). A comprehensive discussion of the ways in which various labile functional groups may be protected and methods for cleaving the resulting protected derivatives is given in, for example, Protective Groups in Organic Chemistry, T.W. Greene and P.G.M. Wuts, (1991) Wiley-Interscience, New York, 2nd edition.
Any novel intermediates, such as those defined above, may be of use in the synthesis of compounds of formula (I) and are therefore also included within the scope of the invention, as well as salts or protected derivatives thereof.
As indicated above the compounds of formula (I) are useful as GPRl 19 agonists, e.g. for the treatment and/or prophylaxis of obesity and diabetes. For such use the compounds of formula (I) will generally be administered in the form of a pharmaceutical composition.
The invention also provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, for use as a pharmaceutical.
The invention also provides a pharmaceutical composition comprising a compound of formula (I), in combination with a pharmaceutically acceptable carrier. Preferably the composition is comprised of a pharmaceutically acceptable carrier and a non-toxic therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
Moreover, the invention also provides a pharmaceutical composition for the treatment of disease by modulating GPRl 19, resulting in the prophylactic or therapeutic treatment of obesity, e.g. by regulating satiety, or for the treatment of diabetes, comprising a pharmaceutically acceptable carrier and a non-toxic therapeutically effective amount of compound of formula (I), or a pharmaceutically acceptable salt thereof.
The pharmaceutical compositions may optionally comprise other therapeutic ingredients or adjuvants. The compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
In practice, the compounds of formula (I), or pharmaceutically acceptable salts thereof, can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g. oral or parenteral (including intravenous).
Thus, the pharmaceutical compositions can be presented as discrete units suitable for oral administration such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient. Further, the compositions can be presented as a powder, as granules, as a solution, as a suspension in an aqueous liquid, as a non-aqueous liquid, as an oil-in- water emulsion, or as a water-in-oil liquid emulsion. In addition to the common dosage forms set out above, the compound of formula (I), or a pharmaceutically acceptable salt thereof, may also be administered by controlled release means and/or delivery devices. The compositions may be prepared by any of the methods of pharmacy. In general, such methods include a step of bringing into association the active ingredient with the carrier that constitutes one or more necessary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both. The product can then be conveniently shaped into the desired presentation.
The compounds of formula (I), or pharmaceutically acceptable salts thereof, can also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds.
The pharmaceutical carrier employed can be, for example, a solid, liquid, or gas. Examples of solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Examples of liquid carriers are sugar syrup, peanut oil, olive oil, and water. Examples of gaseous carriers include carbon dioxide and nitrogen.
In preparing the compositions for oral dosage form, any convenient pharmaceutical media may be employed. For example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like may be used to form oral liquid preparations such as suspensions, elixirs and solutions; while carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be used to form oral solid preparations such as powders, capsules and tablets. Because of their ease of administration, tablets and capsules are the preferred oral dosage units whereby solid pharmaceutical carriers are employed. Optionally, tablets may be coated by standard aqueous or nonaqueous techniques.
A tablet containing the composition of this invention may be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants. Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free- flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent. Each tablet preferably contains from about 0.05mg to about 5g of the active ingredient and each cachet or capsule preferably containing from about 0.05mg to about 5g of the active ingredient.
For example, a formulation intended for the oral administration to humans may contain from about 0.5mg to about 5g of active agent, compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95 percent of the total composition. Unit dosage forms will generally contain between from about lmg to about 2g of the active ingredient, typically 25mg, 50mg, lOOmg, 200mg, 300mg, 400mg, 500mg, 600mg, 800mg, or lOOOmg.
Pharmaceutical compositions of the present invention suitable for parenteral administration may be prepared as solutions or suspensions of the active compounds in water. A suitable surfactant can be included such as, for example, hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms.
Pharmaceutical compositions of the present invention suitable for injectable use include sterile aqueous solutions or dispersions. Furthermore, the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be effectively fluid for easy syringability. The pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g. glycerol, propylene glycol and liquid polyethylene glycol), vegetable oils, and suitable mixtures thereof.
Pharmaceutical compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, or the like. Further, the compositions can be in a form suitable for use in transdermal devices. These formulations may be prepared, using a compound of formula (I), or a pharmaceutically acceptable salt thereof, via conventional processing methods. As an example, a cream or ointment is prepared by admixing hydrophilic material and water, together with about 5wt% to about 10wt% of the compound, to produce a cream or ointment having a desired consistency.
Pharmaceutical compositions of this invention can be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories may be conveniently formed by first admixing the composition with the softened or melted carrier(s) followed by chilling and shaping in molds. In addition to the aforementioned carrier ingredients, the pharmaceutical formulations described above may include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti- oxidants) and the like. Furthermore, other adjuvants can be included to render the formulation isotonic with the blood of the intended recipient. Compositions containing a compound of formula (I), or pharmaceutically acceptable salts thereof, may also be prepared in powder or liquid concentrate form.
Generally, dosage levels on the order of O.Olmg/kg to about 150mg/kg of body weight per day are useful in the treatment of the above- indicated conditions, or alternatively about 0.5mg to about 7g per patient per day. For example, obesity may be effectively treated by the administration of from about 0.01 to 50mg of the compound per kilogram of body weight per day, or alternatively about 0.5mg to about 3.5g per patient per day.
It is understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing therapy.
The compounds of formula (I) may be used in the treatment of diseases or conditions in which GPRl 19 plays a role.
Thus the invention also provides a method for the treatment of a disease or condition in which GPRl 19 plays a role comprising a step of administering to a subject in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof. Diseases or conditions in which GPRl 19 plays a role include obesity and diabetes. In the context of the present application the treatment of obesity is intended to encompass the treatment of diseases or conditions such as obesity and other eating disorders associated with excessive food intake e.g. by reduction of appetite and body weight, maintenance of weight reduction and prevention of rebound and diabetes (including Type 1 and Type 2 diabetes, impaired glucose tolerance, insulin resistance and diabetic complications such as neuropathy, nephropathy, retinopathy, cataracts, cardiovascular complications and dyslipidaemia). And the treatment of patients who have an abnormal sensitivity to ingested fats leading to functional dyspepsia. The compounds of the invention may also be used for treating metabolic diseases such as metabolic syndrome (syndrome X), impaired glucose tolerance, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low HDL levels and hypertension.
The compounds of the invention may offer advantages over compounds acting via different mechanisms for the treatment of the above mentioned disorders in that they may offer beta-cell protection, increased cAMP and insulin secretion and also slow gastric emptying.
The compounds of the invention may also be used for treating conditions characterised by low bone mass such asosteopenia, osteoporosis, rheumatoid arthritis, osteoarthritis, periodontal disease, alveolar bone loss, osteotomy bone loss, childhood idiopathic bone loss, Paget's disease, bone loss due to metastatic cancer, osteolytic lesions, curvature of the spine and loss of height.
The invention also provides a method for the regulation of satiety comprising a step of administering to a subject in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof. The invention also provides a method for the treatment of obesity comprising a step of administering to a subject in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
The invention also provides a method for the treatment of diabetes, including Type 1 and Type 2 diabetes, particularly type 2 diabetes, comprising a step of administering to a patient in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
The invention also provides a method for the treatment of metabolic syndrome (syndrome X), impaired glucose tolerance, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low HDL levels or hypertension comprising a step of administering to a patient in need thereof an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
The invention also provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of a condition as defined above.
The invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a condition as defined above.
In the methods of the invention the term "treatment" includes both therapeutic and prophylactic treatment.
The compounds of formula (I) may exhibit advantageous properties compared to known GPR 119 agonists, for example, the compounds may exhibit improved potency or stability, or improved solubility thus improving absorption properties and bioavailability, or other advantageous properties for compounds to be used as pharmaceuticals.
The compounds of formula (I), or pharmaceutically acceptable salts thereof, may be administered alone or in combination with one or more other therapeutically active compounds. The other therapeutically active compounds may be for the treatment of the same disease or condition as the compounds of formula (I) or a different disease or condition. The therapeutically active compounds may be administered simultaneously, sequentially or separately.
The compounds of formula (I) may be administered with other active compounds for the treatment of obesity and/or diabetes, for example insulin and insulin analogs, gastric lipase inhibitors, pancreatic lipase inhibitors, sulfonyl ureas and analogs, biguanides, α2 agonists, glitazones, PPAR-γ agonists, mixed PPAR-α/γ agonists, RXR agonists, fatty acid oxidation inhibitors, α-glucosidase inhibitors, dipeptidyl peptidase IV inhibitors, GLP-I agonists e.g. GLP-I analogues and mimetics, β-agonists, phosphodiesterase inhibitors, lipid lowering agents, glycogen phosphorylase inhibitors, antiobesity agents e.g. pancreatic lipase inhibitors, MCH-I antagonists and CB-I antagonists (or inverse agonists), amylin antagonists, lipoxygenase inhibitors, somostatin analogs, glucokinase activators, glucagon antagonists, insulin signalling agonists, PTPlB inhibitors, gluconeogenesis inhibitors, antilypolitic agents, GSK inhibitors, galanin receptor agonists, anorectic agents, CCK receptor agonists, leptin, serotonergic/dopaminergic antiobesity drugs, reuptake inhibitors e.g. sibutramine, CRF antagonists, CRF binding proteins, thyromimetic compounds, aldose reductase inhibitors, glucocorticoid receptor antagonists, NHE-I inhibitors or sorbitol dehydrogenase inhibitors. Combination therapy comprising the administration of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and at least one other antiobesity agent represents a further aspect of the invention.
The present invention also provides a method for the treatment of obesity in a mammal, such as a human, which method comprises administering an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and another antiobesity agent, to a mammal in need thereof.
The invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and another antiobesity agent for the treatment of obesity.
The invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for use in combination with another antiobesity agent, for the treatment of obesity.
The compound of formula (I), or a pharmaceutically acceptable salt thereof, and the other antiobesity agent(s) may be co-administered or administered sequentially or separately.
Co-administration includes administration of a formulation which includes both the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the other antiobesity agent(s), or the simultaneous or separate administration of different formulations of each agent. Where the pharmacological profiles of the compound of formula (I), or a pharmaceutically acceptable salt thereof, and the other antiobesity agent(s) allow it, coadministration of the two agents may be preferred.
The invention also provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and another antiobesity agent in the manufacture of a medicament for the treatment of obesity.
The invention also provides a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and another antiobesity agent, and a pharmaceutically acceptable carrier. The invention also encompasses the use of such compositions in the methods described above.
GPRl 19 agonists are of particular use in combination with centrally acting antiobesity agents.
The other antiobesity agent for use in the combination therapies according to this aspect of the invention is preferably a CB-I modulator, e.g. a CB-I antagonist or inverse agonist. Examples of CB-I modulators include SR141716 (rimonabant) and SLV-319 ((45)-(-)-3-(4- chlorophenyl)-N-methyl-N-[(4-chlorophenyl)sulfonyl]-4-phenyl-4,5-dihydro-lH-pyrazole-l- carboxamide); as well as those compounds disclosed in EP576357, EP656354, WO 03/018060, WO 03/020217, WO 03/020314, WO 03/026647, WO 03/026648, WO 03/027076, WO 03/040105, WO 03/051850, WO 03/051851, WO 03/053431, WO 03/063781, WO 03/075660, WO 03/077847, WO 03/078413, WO 03/082190, WO 03/082191, WO 03/082833, WO 03/084930, WO 03/084943, WO 03/086288, WO 03/087037, WO 03/088968, WO 04/012671, WO 04/013120, WO 04/026301, WO 04/029204, WO 04/034968, WO 04/035566, WO 04/037823 WO 04/052864, WO 04/058145, WO 04/058255, WO 04/060870, WO 04/060888, WO 04/069837, WO 04/069837, WO 04/072076, WO 04/072077, WO 04/078261 and WO 04/108728, and the references disclosed therein.
Other diseases or conditions in which GPRl 19 has been suggested to play a role include those described in WO 00/50562 and US 6,468,756, for example cardiovascular disorders, hypertension, respiratory disorders, gestational abnormalities, gastrointestinal disorders, immune disorders, musculoskeletal disorders, depression, phobias, anxiety, mood disorders and Alzheimer's disease.
All publications, including, but not limited to, patents and patent application cited in this specification, are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as fully set forth.
The invention will now be described by reference to the following examples which are for illustrative purposes and are not to be construed as a limitation of the scope of the present invention.
EXAMPLES
Materials and methods
Column chromatography was carried out on Siθ2 (40-63 mesh) unless specified otherwise. LCMS data were obtained as follows: Atlantis 3μ Cig column (3.0 X 20.0 mm, flow rate = 0.85 mL/min) eluting with a H2O-CH3CN solution containing 0.1% HCO2H over 6 min with UV detection at 220 nm. Gradient information: 0.0-0.3 min 100% H2O; 0.3-4.25 min: Ramp up to 10% H2O-90% CH3CN; 4.25-4.4 min: Ramp up to 100% CH3CN; 4.4-4.9 min: Hold at 100% CH3CN; 4.9-6.0 min: Return to 100% H2O. The mass spectra were obtained using an electrospray ionisation source in either the positive (ES+) or negative (ES") ion modes.
Abbreviations and acronyms; Ac: Acetyl; DIPEA: N,N-Diisopropylethylamine; EDCI: 1- (3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride; Et: Ethyl; h: hour(s); HOBt: 1- Hydroxybenzotriazole; IH: Isohexane; /Pr: Isopropyl; Me: Methyl; RT: Retention time; THF: Tetrahydrofuran; TMEDA: N5, N5, N, N-Tetramethylethylenediamine.
The syntheses of the following compounds have been described elsewhere: N- Hydroxyisobutyramidine: J. Org. Chem. 2003, 68, 7316-7321; 3-Piperidin-4-ylpropan-l-ol: Tetrahedron 1999, 55, 11619-11639. All otfher compounds were available from commercial sources.
Preparation 1: 4-(3-Hydroxypropyl)piperidine-l-carbonitrile
Figure imgf000013_0001
A slurry OfNaHCO3 (35.2 g, 0.42 mol) in H2O (70 mL) was added to a stirred solution of 3-piperidin-4-ylpropan-l-ol (20.0 g, 0.14 mol) in CH2Cl2 at 00C. A solution of BrCN (17.8 g, 0.17 mol) in CH2Cl2 (19 mL) was added to the reaction over 1 min, then stirring was continued at 00C for 0.5 h. The reaction was then stirred at 200C for 2 h, before being washed with saturated aqueous NaHCO3 and brine. The CH2Cl2 solution was dried (MgSOzi), filtered and concentrated in vacuo to furnish an oil that was dissolved in a small amount of CH2Cl2, before being filtered through a SiO2 pad, eluting with EtOAc. The filtrate was concentrated under reduced pressure to afford the title compound: mlz (ES+) = 169.1 [M+ H]+.
Preparation 2: 3-[l-(3-Isopropyl[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propan-l-ol
Figure imgf000014_0001
ZnCl2 (IM in Et2O, 145 mL, 145 mmol) was added over 20 min to a stirred solution of 4-(3-hydroxypropyl)piperidine-l-carbonitrile (Preparation 1, 20.3 g, 121 mmol) and N- hydroxyisobutyramidine (14.8 g, 145 mmol) in EtOAc (290 mL) and THF (270 mL). After 2 h, the white precipitate that had formed was collected and washed with THF-EtOAc (1 :1, 50 mL). This precipitate was dissolved in EtOH (550 mL) and 12M HCl (70 mL), then the solution was stirred with heating to 700C for 16 h. The EtOH was removed in vacuo, the remainder was diluted with H2O, then the pH was adjusted to 7 with solid ΝaHCθ3. The mixture was extracted with EtOAc (3x), then the combined extracts were washed with brine, before being dried (MgSOzi). Filtration and solvent removal furnished the title compound: mlz (ES+) = 254.1 [M + H]+.
Preparation 3: 5-{3-[l-(3-Isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propoxy}pyridme-2- carboxylic acid
Figure imgf000014_0002
A stirred solution of methyl 5-hydroxypyridine-2-carboxylate (726 mg, 4.7 mmol), 3-[l- (3-isopropyl[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propan-l-ol (Preparation 2, 1.00 g, 4.0 mmol), and triphenyl phosphine (1.24 g, 4.7 mmol) in THF was treated dropwise with diisopropyl azodicarboxylate (1.1 mL, 4.7 mmol). After 16 h, the reaction was concentrated, then the residue was dissolved in EtOAc. The solution was washed with 2M NaOH (2x) and brine, before being dried (MgSOzi), filtered and concentrated. The remainder was then stirred with a small amount of Et2O at 00C. After 40 min, the mixture was filtered, then the filtrate was concentrated and the residue purified by column chromatography (EtOAc-CH2Cl2, 3:7) to provide methyl 5-{3-[l-(3-isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propoxy}pyridme-2- carboxylate: mlz (ES+) = 389.00 [M + H]+. A mixture of this ester (1.34 g, 3.5 mmol), LiOH-H2O (1.45 g, 34.6 mmol), MeOH (26 mL), and H2O (7 mL) were stirred at 200C for 16 h. The MeOH was removed in vacuo, then more H2O was added. The solution was washed with EtOAc (2x), before being acidified to pH 5 with 2M HCl. The precipitate formed was collected and vacuum-dried to afford the title compound: mlz (ES+) = 374.96 [M+ H]+.
Preparation 4: 5-{3-[l-(3-Isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propoxy}-3-methyl- pyridine-2-carboxylic acid
Figure imgf000014_0003
Mitsunobu condensation of 5-hydroxy-3-methylpyridine-2-carbonitrile (1.00 g, 7.5 mmol) with 3-[l-(3-isopropyl[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propan-l-ol (Preparation 2, 1.57 g, 6.2 mmol), by a procedure similar to that outlined in Preparation 3, provided 5-{3-[l-(3-isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propoxy}-3-methyl- pyridine-2-carbonitrile: mlz (ES+) = 370.01 [M + H]+. A solution of this nitrile (500 mg, 1.4 mmol) in EtOH (8 mL) and 2M NaOH (4 mL, 8.0 mmol) was heated at 700C for 7 h, before being stirred at ambient temperature for 16 h. The EtOH was removed under reduced pressure, then the remainder was diluted with H2O. The solution was washed with EtOAc (2x), before being acidified to pH5 with 2M HCl. The mixture was extracted with EtOAc (2x), then the combined extracts were dried (MgSOzi), filtered and concentrated. The residue was purified by column chromatography (MeOH-CH2Cl2 (1 :9) to yield the title compound: RT = 3.31 min; mlz (ES+) = 389.02 [M + H]+.
Preparation 5: 2-Bromo-5-{3-[l-(3-isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4- yl]propoxy} pyridine
Figure imgf000015_0001
Mitsunobu condensation of 2-bromo-5-hydroxypyridine with 3-[l-(3- isopropyl[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propan-l-ol (Preparation 2), by a procedure similar to that outlined in Preparation 3, provided the title compound: RT = 4.01 min; mlz (ES+) = 409.0 [M+ H]+.
Example 1: 5-{3-[l-(3 -Isopropyl- [ 1 ,2,4]oxadiazol-5 -yl)piperidin-4-yl]propoxy } -3 -methyl- pyridine-2-carboxylic acid ((R)-2-hydroxy-l-methylethyl)amide
Figure imgf000015_0002
A solution of 5-{3-[l-(3 -isopropyl- [ 1 ,2,4]oxadiazol-5 -yl)piperidin-4-yl]propoxy } -3 - methylpyridine-2-carboxylic acid (Preparation 4, 178 mg, 459 μmol), EDCI (176 mg, 917 μmol), HOBt (141 mg, 917 μmol) and DIPEA (160 μL, 917 μmol) in anhydrous THF (8 mL) was stirred for 30 min. (R)-2-Aminopropan-l-ol (108 μL, 1376 μmol) was added, then stirring was continued for 16 h. The solvents were removed under reduced pressure, then CH2Cl2 was added. The solution was washed with H2O, 2M NaOH, H2O and brine, before being dried (MgSOzi). Filtration, solvent evaporation and column chromatography (EtOAc) furnished the title compound: δH (CDCl3) 1.25-1.41 (m, HH), 1.47-1.62 (m, 3H), 1.81-1.96 (m, 4H), 2.77 (s, 3H), 2.93 (sept, IH), 3.02-3.13 (m, 2H), 3.63-3.71 (m, IH), 3.78-3.82 (m, IH), 4.02-4.10 (m, 2H), 4.15-4.28 (m, 3H), 4.45-4.55 (br, IH), 7.02 (d, IH), 8.05-8.15 (m, 2H); RT = 3.49 min; mlz (ES+) = 446.00 [M+ H]+.
Example 2: 5-{3-[l-(3 -Isopropyl- [ 1 ,2,4]oxadiazol-5 -yl)piperidin-4-yl]propoxy } pyridine-2- carboxylic acid ((R)-2-hydroxy-l-methylethyl)amide
Figure imgf000015_0003
Condensation of 5-{3-[l-(3-isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propoxy}- pyridine-2-carboxylic acid (Preparation 3) with (R)-2-aminopropan-l-ol, employing a procedure similar to that used in Example 1, gave the title compound: mlz (ES+) = 432.03 [M + H]+.
Example 3: 5-{3-[l-(3-Isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propoxy}-3- methylpyridine-2-carboxylic acid amide
Figure imgf000016_0001
Condensation of 5-{3-[l-(3-isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4-yl]propoxy}-3- methylpyridine-2-carboxylic acid (Preparation 4) with ammonia, employing a procedure similar to that used in Example 1, gave the title compound: RT = 3.62 min; mlz (ES+) = 388.02 [M + H]+.
Example 4: 5-{3-[l-(3 -Isopropyl- [ 1 ,2,4]oxadiazol-5 -yl)piperidin-4-yl]propoxy } -2- methanesulfonylpyridine
Figure imgf000016_0002
W-BuLi (322 μL of a 2.5 M solution in hexanes, 0.81 mmol) was added dropwise to a stirred solution of 2-bromo-5-{3-[l-(3-isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4- yl]propoxy} pyridine (Preparation 5, 300 mg, 0.73 mmol) in anhydrous THF (4 mL) at -78°C. After 15 min, the reaction was treated with (MeS)2 (132 μL, 1.47 mmol), then stirring was continued at -78°C for an additional 15 min. The reaction was then allowed to warm to -500C over 1 h, before finally being warmed up to room temperature. The mixture was partitioned between IM HCl and EtOAc, then the organic phase was washed with IM NaOH and brine, before being dried (MgSOzi). Filtration, solvent evaporation and column chromatography (EtOAc-IH, 15% to 30%) afforded 5-{3-[l-(3-isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4- yl]propoxy}-2-methylsulfanylpyridine. A stirred solution of this thioether (75 mg, 0.2 mmol) in CH2CI2 (10 mL) was treated with 3-chloroperbenzoic acid (99 mg of 70% pure, 0.4 mmol). After 40 min, the reaction was quenched with aqueous Na2Cθ3, then the organic phase was separated and concentrated. The residue was purified by column chromatography (EtOAc-IH, 20% to 50%) to yield the title compound: RT = 3.55 min; mlz (ES+) = 409.18 [M+ H]+.
The following compound may also be made by the processes described above: 5-{(R)-3-[l-(3-Isopropyl-[l,2,4]oxadiazol-5-yl)piperidin-4-yl]butoxy}-2- methanesulfonylpyridine
The biological activity of the compounds of the invention may be tested in the following assay systems: Yeast Reporter Assay
The yeast cell-based reporter assays have previously been described in the literature (e.g. see Miret J. J. et al, 2002, J. Biol. Chem., 277:6881-6887; Campbell R.M. et al, 1999, Bioorg. Med. Chem. Lett, 9:2413-2418; King K. et al, 1990, Science, 250:121-123); WO 99/14344; WO 00/12704; and US 6,100,042). Briefly, yeast cells have been engineered such that the endogenous yeast G-alpha (GPAl) has been deleted and replaced with G-protein chimeras constructed using multiple techniques. Additionally, the endogenous yeast GPCR, Ste3 has been deleted to allow for heterologous expression of a mammalian GPCR of choice. In the yeast, elements of the pheromone signaling transduction pathway, which are conserved in eukaryotic cells (for example, the mitogen-activated protein kinase pathway), drive the expression of Fusl. By placing β-galactosidase (LacZ) under the control of the Fusl promoter (Fuslp), a system has been developed whereby receptor activation leads to an enzymatic readout.
Yeast cells were transformed by an adaptation of the lithium acetate method described by Agatep et al, (Agatep, R. et al, 1998, Transformation of Saccharomyces cerevisiae by the lithium acetate/single-stranded carrier DNA/polyethylene glycol (LiAc/ss-DNA/PEG) protocol. Technical Tips Online, Trends Journals, Elsevier). Briefly, yeast cells were grown overnight on yeast tryptone plates (YT). Carrier single-stranded DNA (lOμg), 2μg of each of two Fuslp- LacZ reporter plasmids (one with URA selection marker and one with TRP), 2μg of GPRl 19 (human or mouse receptor) in yeast expression vector (2μg origin of replication) and a lithium acetate/ polyethylene glycol/ TE buffer was pipetted into an Eppendorf tube. The yeast expression plasmid containing the receptor/ no receptor control has a LEU marker. Yeast cells were inoculated into this mixture and the reaction proceeds at 300C for 60min. The yeast cells were then heat-shocked at 42°C for 15min. The cells were then washed and spread on selection plates. The selection plates are synthetic defined yeast media minus LEU, URA and TRP (SD- LUT). After incubating at 300C for 2-3 days, colonies that grow on the selection plates were then tested in the LacZ assay.
In order to perform fluorimetric enzyme assays for β-galactosidase, yeast cells carrying the human or mouse GPRl 19 receptor were grown overnight in liquid SD-LUT medium to an unsaturated concentration (i.e. the cells were still dividing and had not yet reached stationary phase). They were diluted in fresh medium to an optimal assay concentration and 90μl of yeast cells added to 96-well black polystyrene plates (Costar). Compounds, dissolved in DMSO and diluted in a 10% DMSO solution to 1OX concentration, were added to the plates and the plates placed at 300C for 4h. After 4h, the substrate for the β-galactosidase was added to each well. In these experiments, Fluorescein di (β-D-galactopyranoside) was used (FDG), a substrate for the enzyme that releases fluorescein, allowing a fluorimetric read-out. 20μl per well of 500μM FDG/2.5% Triton XlOO was added (the detergent was necessary to render the cells permeable). After incubation of the cells with the substrate for 60min, 20μl per well of IM sodium carbonate was added to terminate the reaction and enhance the fluorescent signal. The plates were then read in a fluorimeter at 485/535nm.
The compounds of the invention give an increase in fluorescent signal of at least ~ 1.5- fold that of the background signal (i.e. the signal obtained in the presence of 1% DMSO without compound). Compounds of the invention which give an increase of at least 5-fold may be preferred.
cAMP Assay
A stable cell line expressing recombinant human GPRl 19 was established and this cell line may be used to investigate the effect of compounds of the invention on intracellular levels of cyclic AMP (cAMP). The cell monolayers are washed with phosphate buffered saline and stimulated at 37°C for 30min with various concentrations of compound in stimulation buffer plus 1% DMSO. Cells are then lysed and cAMP content determined using the Perkin Elmer AlphaScreen™ (Amplified Luminescent Proximity Homogeneous Assay) cAMP kit. Buffers and assay conditions are as described in the manufacturer's protocol.
In vivo feeding study
The effect of compounds of the invention on body weight and food and water intake may be examined in freely- feeding male Sprague-Dawley rats maintained on reverse-phase lighting. Test compounds and reference compounds are dosed by appropriate routes of administration (e.g. intraperitoneally or orally) and measurements made over the following 24 h. Rats are individually housed in polypropylene cages with metal grid floors at a temperature of 21±4°C and 55±20% humidity. Polypropylene trays with cage pads are placed beneath each cage to detect any food spillage. Animals are maintained on a reverse phase light-dark cycle (lights off for 8 h from 09.30-17.30 h) during which time the room is illuminated by red light. Animals have free access to a standard powdered rat diet and tap water during a two week acclimatization period. The diet is contained in glass feeding jars with aluminum lids. Each lid has a 3-4 cm hole in it to allow access to the food. Animals, feeding jars and water bottles are weighed (to the nearest 0.1 g) at the onset of the dark period. The feeding jars and water bottles are subsequently measured 1, 2, 4, 6 and 24 h after animals are dosed with a compound of the invention and any significant differences between the treatment groups at baseline compared to vehicle-treated controls.
Anti-diabetic effects of compounds of the invention in an in-vitro model of pancreatic beta cells (HIT-T15)
Cell Culture
HIT-Tl 5 cells (passage 60) were obtained from ATCC, and were cultured in RPMI 1640 medium supplemented with 10% fetal calf serum and 3OnM sodium selenite. All experiments were done with cells at less than passage 70, in accordance with the literature, which describes altered properties of this cell line at passage numbers above 81 (Zhang HJ, Walseth TF, Robertson RP. Insulin secretion and cAMP metabolism in HIT cells. Reciprocal and serial passage-dependent relationships. Diabetes. 1989 Jan;38(l):44-8).
cAMP assay
HIT-T15 cells were plated in standard culture medium in 96-well plates at 100,000 cells/ 0.1ml/ well and cultured for 24 hr and the medium was then discarded. Cells were incubated for 15min at room temperature with lOOμl stimulation buffer (Hanks buffered salt solution, 5mM HEPES, 0.5mM IBMX, 0.1% BSA, pH 7.4). This was discarded and replaced with compound dilutions over the range 0.001, 0.003, 0.01, 0.03, 0.1, 0.3, 1, 3, 10, 30 μM in stimulation buffer in the presence of 0.5% DMSO. Cells were incubated at room temperature for 30min. Then 75ul lysis buffer (5mM HEPES, 0.3% Tween-20, 0.1% BSA, pH 7.4) was added per well and the plate was shaken at 900 rpm for 20 min. Particulate matter was removed by centrifugation at 3000rpm for 5min, then the samples were transferred in duplicate to 384-well plates, and processed following the Perkin Elmer AlphaScreen cAMP assay kit instructions. Briefly 25μl reactions were set up containing 8μl sample, 5μl acceptor bead mix and 12μl detection mix, such that the concentration of the final reaction components is the same as stated in the kit instructions. Reactions were incubated at room temperature for 150min, and the plate was read using a Packard Fusion instrument. Measurements for cAMP were compared to a standard curve of known cAMP amounts (0.01, 0.03, 0.1, 0.3, 1, 3, 10, 30, 100, 300, 1000 nM) to convert the readings to absolute cAMP amounts. Data was analysed using XLfit 3 software.
Representative compounds of the invention were found to increase cAMP at an EC5O of less than 10 μM. Compounds showing an EC5O of less than 1 μM in the cAMP assay may be preferred.
Insulin secretion assay
HIT-Tl 5 cells are plated in standard culture medium in 12-well plates at 106 cells/ 1 ml/ well and cultured for 3 days and the medium is then discarded. Cells are washed x 2 with supplemented Krebs-Ringer buffer (KRB) containing 119 mM NaCl, 4.74 mM KCl, 2.54 mM CaCl2, 1.19 mM MgSO4, 1.19 mM KH2PO4, 25 mM NaHCO3, 1OmM HEPES at pH 7.4 and 0.1% bovine serum albumin. Cells are incubated with ImI KRB at 37°C for 30 min which is then discarded. This is followed by a second incubation with KRB for 30 min, which is collected and used to measure basal insulin secretion levels for each well. Compound dilutions (0, 0.1, 0.3, 1, 3, 10 uM) are then added to duplicate wells in ImI KRB, supplemented with 5.6 mM glucose. After 30 min incubation at 37°C samples are removed for determination of insulin levels. Measurement of insulin is done using the Mercodia Rat insulin ELISA kit, following the manufacturers instructions, with a standard curve of known insulin concentrations. For each well insulin levels are corrected by subtraction of the basal secretion level from the preincubation in the absence of glucose. Data is analysed using XLfit 3 software.
Oral Glucose Tolerance Tests
The effects of compounds of the invention on oral glucose (GIc) tolerance were evaluated in male Sprague-Dawley rats. Food was withdrawn 16 h before administration of GIc and remained withdrawn throughout the study. Rats had free access to water during the study. A cut was made to the animals' tails, then blood (1 drop) was removed for measurement of basal GIc levels 60 min before administration of the GIc load. Then, the rats were weighed and dosed orally with test compound or vehicle (20% aqueous hydroxypropyl-/S-cyclodextrin) 45 min before the removal of an additional blood sample and treatment with the GIc load (2 g kg"1 p.o.). Blood samples were then taken from the cut tip of the tail 5, 15, 30, 60, 120, and 180 min after GIc administration. Blood glucose levels were measured just after collection using a commercially available glucose-meter (OneTouch® UltraTM from Lifescan). Representative compounds of the invention statistically reduced the GIc excursion at doses of <10 mg kg"1.
The effects of compounds of the invention on oral glucose (GIc) tolerance may also be evaluated in male C57B1/6 or male oblob mice. Food is withdrawn 5 h before administration of GIc and remains withdrawn throughout the study. Mice have free access to water during the study. A cut is made to the animals' tails, then blood (20 μL) is removed for measurement of basal GIc levels 45 min before administration of the GIc load. Then, the mice are weighed and dosed orally with test compound or vehicle (20% aqueous hydroxypropyl-yS-cyclodextrin or 25% aqueous Gelucire 44/14) 30 min before the removal of an additional blood sample (20 μL) and treatment with the GIc load (2-5 g kg"1 p.o.). Blood samples (20 μL) are then taken 25, 50, 80, 120, and 180 min after GIc administration. The 20 μL blood samples for measurement of GIc levels are taken from the cut tip of the tail into disposable micro-pipettes (Dade Diagnostics Inc., Puerto Rico) and the sample added to 480 μL of haemolysis reagent. Duplicate 20 μL aliquots of the diluted haemolysed blood are then added to 180 μL of Trinders glucose reagent (Sigma enzymatic (Trinder) colorimetric method) in a 96-well assay plate. After mixing, the samples are left at rt for 30 min before being read against GIc standards (Sigma glucose/urea nitrogen combined standard set).

Claims

WHAT IS CLAIMED IS:
1. A compound of formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000021_0001
(I) wherein one of X and Y is O and the other is N;
R1 is SO2R5, NR6R7, or -CONR6R7;
R2 is hydrogen or methyl;
R3 is hydrogen or methyl;
R4 is C2-5 alkyl;
R5 is Ci_3alkyl;
R6 and R7 are independently hydrogen, C 1.4 alkyl, which may optionally be substituted by halo, hydroxy,
Figure imgf000021_0002
alkoxy-, aryloxy-,
Figure imgf000021_0003
alkoxy-, Ci_4 alkylS(0)m-, C3.7 heterocyclyl, N(R8)2 or -C(O)OR9; or may be C3.7 cycloalkyl, aryl, heterocyclyl or heteroaryl, wherein the cyclic groups may be substituted with one or more substituents selected from halo, C 1.4 alkyl, Ci- 4 fluoroalkyl, OR6, CN, SO2CH3, N(R8)2 and NO2; or taken together R6 and R7 may form a 5- or 6-membered heterocyclic ring optionally substituted by hydroxy, C 1.4 alkyl or C 1.4 hydroxyalkyl and optionally containing a further heteroatom selected from O and NR8;
R8 are independently hydrogen or C 1.4 alkyl; or a group N(R8)2 may form a 4- to 7- membered heterocyclic ring optionally containing a further heteroatom selected from O and NR8;
R9 is hydrogen or C 1.4 alkyl; and m is O, 1 or 2.
2. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein X is O.
3. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein Y is O.
4. A compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R1 is SO2R5.
5. A compound according to claim 4, or a pharmaceutically acceptable salt thereof, wherein R1 is SO2CH3.
6. A compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R1 is -CONR6R7.
7. A compound according to claim 6, or a pharmaceutically acceptable salt thereof, wherein R6 is hydrogen, Ci_3alkyl, or C2-3alkyl substituted by hydroxy and R7 is hydrogen.
8. A compound according to claim 7, or a pharmaceutically acceptable salt thereof, wherein R6 is C^alkyl or C2-3alkyl substituted by hydroxy.
9. A compound according to claim 8, or a pharmaceutically acceptable salt thereof, wherein R6 is C2-3alkyl substituted by hydroxy.
10. A compound according to claim 9, or a pharmaceutically acceptable salt thereof, wherein R6 is 2 -hydroxy ethyl or 2-hydroxy-l-methylethyl.
11. A compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, wherein R3 is hydrogen.
12. A compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt thereof, wherein R3 is methyl.
13. A compound according to claim 12, or a pharmaceutically acceptable salt thereof, wherein R3 is methyl and the sterocentre produced has the (R)-configuration.
14. A compound according to any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein R4 is C3.4 alkyl,
15. A compound according to claim 14, or a pharmaceutically acceptable salt thereof, wherein R4 is isopropyl.
16. A compound of formula (I) as defined in any one of Examples 1 to 4, or a pharmaceutically acceptable salt thereof.
17. A pharmaceutical composition comprising a compound according to any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
18. A method for the treatment of a disease or condition in which GPRl 19 plays a role comprising a step of administering to a subject in need thereof an effective amount of a compound according to any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof.
19. A method for the regulation of satiety comprising a step of administering to a subject in need thereof an effective amount of a compound according to any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof.
20. A method for the treatment of obesity comprising a step of administering to a subject in need thereof an effective amount of a compound according to any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof.
21. A method for the treatment of diabetes comprising a step of administering to a subject in need thereof an effective amount of a compound according to any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof.
22. A method for the treatment of metabolic syndrome (syndrome X), impaired glucose tolerance, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low HDL levels or hypertension comprising a step of administering to a patient in need thereof an effective amount of a compound according to any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof.
23. A compound according to any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof, for use as a medicament.
24. A compound according to any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof, for use in the manufacture of a medicament for the treatment or prevention of a disease or condition as defined in any one of claims 18 to 22.
25. A compound according to any one of claims 1 to 16 or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a disease or condition as defined in any one of claims 17 to 21.
PCT/GB2008/050014 2007-01-04 2008-01-04 Piperidine gpcr agonists WO2008081208A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
BRPI0806312-5A BRPI0806312A2 (en) 2007-01-04 2008-01-04 cgpr piperidine agonists
JP2009544451A JP2010514832A (en) 2007-01-04 2008-01-04 Piperidine GPCR agonist
EA200900878A EA016507B1 (en) 2007-01-04 2008-01-04 Piperidine gpcr agonists
US12/522,030 US20100048625A1 (en) 2007-01-04 2008-01-04 Piperidine gpcr agonists
EP08702097A EP2114936A1 (en) 2007-01-04 2008-01-04 Piperidine gpcr agonists
CA002674360A CA2674360A1 (en) 2007-01-04 2008-01-04 Piperidine gpcr agonists

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
GB0700126.6 2007-01-04
GB0700126A GB0700126D0 (en) 2007-01-04 2007-01-04 GPCR agonists
GB0708748.9 2007-05-08
GB0708748A GB0708748D0 (en) 2007-05-08 2007-05-08 GPCR agonists
GB0719765.0 2007-10-10
GB0719765A GB0719765D0 (en) 2007-10-10 2007-10-10 GPCR agonists

Publications (1)

Publication Number Publication Date
WO2008081208A1 true WO2008081208A1 (en) 2008-07-10

Family

ID=39186140

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2008/050014 WO2008081208A1 (en) 2007-01-04 2008-01-04 Piperidine gpcr agonists

Country Status (7)

Country Link
US (1) US20100048625A1 (en)
EP (1) EP2114936A1 (en)
JP (1) JP2010514832A (en)
BR (1) BRPI0806312A2 (en)
CA (1) CA2674360A1 (en)
EA (1) EA016507B1 (en)
WO (1) WO2008081208A1 (en)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009034388A1 (en) 2007-09-10 2009-03-19 Prosidion Limited Compounds for the treatment of metabolic disorders
WO2010004347A1 (en) * 2008-07-10 2010-01-14 Prosidion Limited Heterocyclic gpcr agonists
WO2010004345A1 (en) * 2008-07-10 2010-01-14 Prosidion Limited Piperidinyl gpcr agonists
WO2010004348A1 (en) * 2008-07-10 2010-01-14 Prosidion Limited Heterocyclic gpcr agonists
WO2010103333A1 (en) 2009-03-12 2010-09-16 Prosidion Limited Compounds for the treatment of metabolic disorders
WO2010103334A1 (en) 2009-03-12 2010-09-16 Prosidion Limited Compounds for the treatment of metabolic disorders
WO2010103335A1 (en) 2009-03-12 2010-09-16 Prosidion Limited Compounds for the treatment of metabolic disorders
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
US20110230507A1 (en) * 2008-07-10 2011-09-22 Prosidion Limited Piperidine GPCR Agonists
WO2011113947A1 (en) 2010-03-18 2011-09-22 Boehringer Ingelheim International Gmbh Combination of a gpr119 agonist and the dpp-iv inhibitor linagliptin for use in the treatment of diabetes and related conditions
WO2011128394A1 (en) 2010-04-14 2011-10-20 Prosidion Limited 3-substituted 5-(pyrrolidine-1-carbonyl) pyrrolidine and its derivatives for use in the treatment of metabolic disorders
WO2011128395A1 (en) 2010-04-14 2011-10-20 Prosidion Limited N- substituted 3-amino 4 - ( pyrrolidine - 1 - carbonyl) pyrrolidine and its derivatives for use in the treatment of metabolic disorders
WO2011147951A1 (en) 2010-05-28 2011-12-01 Prosidion Limited Cycloamino derivatives as gpr119 antagonists
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2012066077A1 (en) 2010-11-18 2012-05-24 Prosidion Limited 1,4 di substituted pyrrolidine - 3 - yl -amine derivatives and their use for the treatment of metabolic disorders
WO2012101292A1 (en) * 2011-01-25 2012-08-02 Viviabiotech, S.L. 1,2,4-oxadiazole derivatives as drugs modulating the glp-1 peptide receptor
GB2488360A (en) * 2011-02-25 2012-08-29 Prosidion Ltd Heterocyclic GPCR agonists
US8293729B2 (en) 2009-06-24 2012-10-23 Boehringer Ingelheim International Gmbh Compounds, pharmaceutical composition and methods relating thereto
WO2012170867A1 (en) 2011-06-09 2012-12-13 Rhizen Pharmaceuticals Sa Novel compounds as modulators of gpr-119
WO2013026587A1 (en) 2011-08-22 2013-02-28 Prosidion Limited 1,4 disubstituted pyrrolidine - 3 - yl -amine derivatives and their use for the treatment of metabolic disorders
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
US8481731B2 (en) 2009-06-24 2013-07-09 Boehringer Ingelheim International Gmbh Compounds, pharmaceutical composition and methods relating thereto

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PE20081849A1 (en) * 2007-01-04 2009-01-26 Prosidion Ltd PIPERIDIN-4-IL-PROPOXY-BENZAMIDE DERIVATIVES AS GPCR AGONISTS

Citations (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0576357A1 (en) 1992-06-23 1993-12-29 Sanofi Pyrazole derivatives, process for their preparation and pharmaceutical compositions containing them
EP0656354A1 (en) 1993-12-02 1995-06-07 Sanofi Substituted N-piperidino 3-pyrazolecarboxamide
WO2000050562A2 (en) 1999-02-22 2000-08-31 Synaptic Pharmaceutical Corporation Dna encoding snorf25 receptor
WO2003018060A1 (en) 2001-08-29 2003-03-06 Aventis Pharma S.A. Composition for treating parkinson's disease containing a cb1 receptor antagonist and a product activating dopaminergic neurotransmission in the brain
WO2003020217A2 (en) 2001-08-31 2003-03-13 University Of Connecticut Novel pyrazole analogs acting on cannabinoid receptors
WO2003020314A1 (en) 2001-08-29 2003-03-13 Aventis Pharma S.A. Compositions for the treatment of parkinson's disease containing a cb1 receptor antagonist and a product that activates dopaminergic neurotransmission in the brain
WO2003026647A1 (en) 2001-09-21 2003-04-03 Solvay Pharmaceuticals B.V. Novel 4,5-dihydro-1h-pyrazole derivatives having cb1-antagonistic activity
WO2003026648A1 (en) 2001-09-21 2003-04-03 Solvay Pharmaceuticals B.V. 4,5-dihydro-1h-pyrazole derivatives having potent cb1-antagonistic activity
WO2003027076A2 (en) 2001-09-21 2003-04-03 Solvay Pharmaceuticals B.V. 1h-imidazole derivatives having cb1 agonistic, cb1 partial agonistic or cb1- antagonistic activity
WO2003040105A1 (en) 2001-11-08 2003-05-15 Sanofi-Synthelabo Polymorphous form of rimonabant, preparation method and pharmaceutical compositions containing same
WO2003051850A1 (en) 2001-12-19 2003-06-26 Astrazeneca Ab Pyrazine compounds and pharmaceutical compositions containing them
WO2003051851A1 (en) 2001-12-19 2003-06-26 Astrazeneca Ab 5, 6-diaryl-pyrazine-2-amide derivatives as cb1 antagonists
WO2003053431A2 (en) 2001-12-21 2003-07-03 Aventis Pharma S.A. Pharmaceutical compositions based on azetidine derivatives
WO2003063781A2 (en) 2002-01-29 2003-08-07 Merck & Co., Inc. Substituted imidazoles as cannabinoid receptor modulators
WO2003075660A1 (en) 2002-03-06 2003-09-18 Merck & Co., Inc. Method of treatment or prevention of obesity
WO2003078413A1 (en) 2002-03-18 2003-09-25 Solvay Pharmaceuticals B.V. Thiazole derivatives having cb1-antagonistic, agonistic or partial agonistic activity
WO2003077847A2 (en) 2002-03-12 2003-09-25 Merck & Co., Inc. Substituted amides
WO2003082190A2 (en) 2002-03-26 2003-10-09 Merck & Co., Inc. Spirocyclic amides as cannabinoid receptor modulators
WO2003082191A2 (en) 2002-03-28 2003-10-09 Merck & Co., Inc. Substituted 2,3-diphenyl pyridines
WO2003082833A1 (en) 2002-03-27 2003-10-09 Consejo Superior De Investigaciones Científicas 1,2,4-triazole derivatives with cannabinoid properties
WO2003084943A2 (en) 2002-04-11 2003-10-16 Sanofi-Aventis Terphenyl derivatives, preparation thereof, compositions containing same
WO2003084930A1 (en) 2002-04-11 2003-10-16 Sanofi-Synthelabo Diphenylpyridine derivatives, the preparation thereof, and pharmaceutical compositions containing said derivatives
WO2003087037A1 (en) 2002-04-05 2003-10-23 Merck & Co., Inc. Substituted aryl amides
WO2003086288A2 (en) 2002-04-12 2003-10-23 Merck & Co., Inc. Bicyclic amides
WO2003088968A1 (en) 2002-04-15 2003-10-30 Research Triangle Institute Compounds having unique cb1 receptor binding selectivity and methods for their production and use
WO2004012671A2 (en) 2002-08-02 2004-02-12 Merck & Co., Inc. Substituted furo [2,3-b] pyridine derivatives
WO2004013120A1 (en) 2002-07-29 2004-02-12 F. Hoffmann-La Roche Ag Novel benzodioxoles
WO2004026301A1 (en) 2002-09-19 2004-04-01 Solvay Pharmaceuticals B.V. 1h-1,2,4-triazole-3-carboxamide derivatives as cannabinoid-cb1 receptor ligands
WO2004029204A2 (en) 2002-09-27 2004-04-08 Merck & Co., Inc. Substituted pyrimidines
WO2004035566A1 (en) 2002-10-18 2004-04-29 Pfizer Products Inc. Cannabinoid receptor ligands and uses thereof
WO2004034968A2 (en) 2002-08-20 2004-04-29 The Regents Of The University Of California Combination therapy for controlling appetites
WO2004037823A1 (en) 2002-10-28 2004-05-06 Pfizer Products Inc. Purine compounds and uses thereof as cannabinoid receptor ligands
WO2004052864A1 (en) 2002-12-12 2004-06-24 Pfizer Products Inc. Pyrazole and imidazole compounds and uses thereof
WO2004058255A1 (en) 2002-12-24 2004-07-15 Astrazeneca Ab 4, 5-diarylthiazole derivatives as cb-1 ligands
WO2004058145A2 (en) 2002-12-19 2004-07-15 Merck & Co., Inc. Substituted amides
WO2004060888A1 (en) 2003-01-02 2004-07-22 F. Hoffmann-La Roche Ag Novel cb 1 receptor inverse agonists
WO2004060870A1 (en) 2003-01-02 2004-07-22 F. Hoffmann-La Roche Ag Novel cb 1 receptour inverse agonists
WO2004069837A1 (en) 2003-02-06 2004-08-19 Pfizer Products Inc. Pyrazolo`1,5-a!`1,3,5!triazine derivatives as cannabinoid receptor ligands
WO2004072077A1 (en) 2003-02-13 2004-08-26 Aventis Pharma Deutshland Gmbh Nitrogen-substituted hexahydropyrazino[1,2-a]pyrimidine-4,7-dione derivatives, method for the production and use thereof as medicaments
WO2004072076A1 (en) 2003-02-13 2004-08-26 Aventis Pharma Deutschland Gmbh Substituted hexahydropyrazino(1,2-a)pyrimidin-4,7-dion derivatives, method for the production and use thereof as medicaments
WO2004078261A1 (en) 2003-03-07 2004-09-16 The University Court Of The University Of Aberdeen Cannabinoid receptor inverse agonists and neutral antagonists as therapeutic agents for the treatment of bone disorders
WO2004108728A1 (en) 2003-06-09 2004-12-16 Pfizer Products Inc. Cannabinoid receptor ligands and uses thereof
WO2005061489A1 (en) 2003-12-24 2005-07-07 Prosidion Limited Heterocyclic derivatives as gpcr receptor agonists
WO2006067531A1 (en) 2004-12-24 2006-06-29 Prosidion Ltd G-protein coupled receptor (gpr116) agonists and use thereof for treating obesity and diabetes
WO2006067532A1 (en) 2004-12-24 2006-06-29 Prosidion Ltd G-protein coupled receptor agonists
WO2006070208A1 (en) 2004-12-31 2006-07-06 Prosidion Ltd. Pyridine, pyrimidine and pyrazine derivatives as gpcr agonists
WO2007003961A2 (en) 2005-06-30 2007-01-11 Prosidion Limited Gpcr agonists
WO2007003964A1 (en) 2005-06-30 2007-01-11 Prosidion Limited G-protein coupled receptor agonists
WO2007003962A2 (en) 2005-06-30 2007-01-11 Prosidion Limited Gpcr agonists
WO2007003960A1 (en) 2005-06-30 2007-01-11 Prosidion Limited Gpcr agonists
WO2007116229A1 (en) * 2006-04-06 2007-10-18 Prosidion Limited Heterocyclic gpcr agonists

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6100042A (en) * 1993-03-31 2000-08-08 Cadus Pharmaceutical Corporation Yeast cells engineered to produce pheromone system protein surrogates, and uses therefor
CN101657471B (en) * 2006-12-06 2013-07-03 史密丝克莱恩比彻姆公司 Bicyclic compounds and use as antidiabetics

Patent Citations (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0576357A1 (en) 1992-06-23 1993-12-29 Sanofi Pyrazole derivatives, process for their preparation and pharmaceutical compositions containing them
EP0656354A1 (en) 1993-12-02 1995-06-07 Sanofi Substituted N-piperidino 3-pyrazolecarboxamide
WO2000050562A2 (en) 1999-02-22 2000-08-31 Synaptic Pharmaceutical Corporation Dna encoding snorf25 receptor
US6468756B1 (en) 1999-02-22 2002-10-22 Synaptic Pharmaceutical Corporation Methods of identifying compounds that bind to SNORF25 receptors
WO2003018060A1 (en) 2001-08-29 2003-03-06 Aventis Pharma S.A. Composition for treating parkinson's disease containing a cb1 receptor antagonist and a product activating dopaminergic neurotransmission in the brain
WO2003020314A1 (en) 2001-08-29 2003-03-13 Aventis Pharma S.A. Compositions for the treatment of parkinson's disease containing a cb1 receptor antagonist and a product that activates dopaminergic neurotransmission in the brain
WO2003020217A2 (en) 2001-08-31 2003-03-13 University Of Connecticut Novel pyrazole analogs acting on cannabinoid receptors
WO2003026647A1 (en) 2001-09-21 2003-04-03 Solvay Pharmaceuticals B.V. Novel 4,5-dihydro-1h-pyrazole derivatives having cb1-antagonistic activity
WO2003026648A1 (en) 2001-09-21 2003-04-03 Solvay Pharmaceuticals B.V. 4,5-dihydro-1h-pyrazole derivatives having potent cb1-antagonistic activity
WO2003027076A2 (en) 2001-09-21 2003-04-03 Solvay Pharmaceuticals B.V. 1h-imidazole derivatives having cb1 agonistic, cb1 partial agonistic or cb1- antagonistic activity
WO2003040105A1 (en) 2001-11-08 2003-05-15 Sanofi-Synthelabo Polymorphous form of rimonabant, preparation method and pharmaceutical compositions containing same
WO2003051851A1 (en) 2001-12-19 2003-06-26 Astrazeneca Ab 5, 6-diaryl-pyrazine-2-amide derivatives as cb1 antagonists
WO2003051850A1 (en) 2001-12-19 2003-06-26 Astrazeneca Ab Pyrazine compounds and pharmaceutical compositions containing them
WO2003053431A2 (en) 2001-12-21 2003-07-03 Aventis Pharma S.A. Pharmaceutical compositions based on azetidine derivatives
WO2003063781A2 (en) 2002-01-29 2003-08-07 Merck & Co., Inc. Substituted imidazoles as cannabinoid receptor modulators
WO2003075660A1 (en) 2002-03-06 2003-09-18 Merck & Co., Inc. Method of treatment or prevention of obesity
WO2003077847A2 (en) 2002-03-12 2003-09-25 Merck & Co., Inc. Substituted amides
WO2003078413A1 (en) 2002-03-18 2003-09-25 Solvay Pharmaceuticals B.V. Thiazole derivatives having cb1-antagonistic, agonistic or partial agonistic activity
WO2003082190A2 (en) 2002-03-26 2003-10-09 Merck & Co., Inc. Spirocyclic amides as cannabinoid receptor modulators
WO2003082833A1 (en) 2002-03-27 2003-10-09 Consejo Superior De Investigaciones Científicas 1,2,4-triazole derivatives with cannabinoid properties
WO2003082191A2 (en) 2002-03-28 2003-10-09 Merck & Co., Inc. Substituted 2,3-diphenyl pyridines
WO2003087037A1 (en) 2002-04-05 2003-10-23 Merck & Co., Inc. Substituted aryl amides
WO2003084943A2 (en) 2002-04-11 2003-10-16 Sanofi-Aventis Terphenyl derivatives, preparation thereof, compositions containing same
WO2003084930A1 (en) 2002-04-11 2003-10-16 Sanofi-Synthelabo Diphenylpyridine derivatives, the preparation thereof, and pharmaceutical compositions containing said derivatives
WO2003086288A2 (en) 2002-04-12 2003-10-23 Merck & Co., Inc. Bicyclic amides
WO2003088968A1 (en) 2002-04-15 2003-10-30 Research Triangle Institute Compounds having unique cb1 receptor binding selectivity and methods for their production and use
WO2004013120A1 (en) 2002-07-29 2004-02-12 F. Hoffmann-La Roche Ag Novel benzodioxoles
WO2004012671A2 (en) 2002-08-02 2004-02-12 Merck & Co., Inc. Substituted furo [2,3-b] pyridine derivatives
WO2004034968A2 (en) 2002-08-20 2004-04-29 The Regents Of The University Of California Combination therapy for controlling appetites
WO2004026301A1 (en) 2002-09-19 2004-04-01 Solvay Pharmaceuticals B.V. 1h-1,2,4-triazole-3-carboxamide derivatives as cannabinoid-cb1 receptor ligands
WO2004029204A2 (en) 2002-09-27 2004-04-08 Merck & Co., Inc. Substituted pyrimidines
WO2004035566A1 (en) 2002-10-18 2004-04-29 Pfizer Products Inc. Cannabinoid receptor ligands and uses thereof
WO2004037823A1 (en) 2002-10-28 2004-05-06 Pfizer Products Inc. Purine compounds and uses thereof as cannabinoid receptor ligands
WO2004052864A1 (en) 2002-12-12 2004-06-24 Pfizer Products Inc. Pyrazole and imidazole compounds and uses thereof
WO2004058145A2 (en) 2002-12-19 2004-07-15 Merck & Co., Inc. Substituted amides
WO2004058255A1 (en) 2002-12-24 2004-07-15 Astrazeneca Ab 4, 5-diarylthiazole derivatives as cb-1 ligands
WO2004060888A1 (en) 2003-01-02 2004-07-22 F. Hoffmann-La Roche Ag Novel cb 1 receptor inverse agonists
WO2004060870A1 (en) 2003-01-02 2004-07-22 F. Hoffmann-La Roche Ag Novel cb 1 receptour inverse agonists
WO2004069837A1 (en) 2003-02-06 2004-08-19 Pfizer Products Inc. Pyrazolo`1,5-a!`1,3,5!triazine derivatives as cannabinoid receptor ligands
WO2004072077A1 (en) 2003-02-13 2004-08-26 Aventis Pharma Deutshland Gmbh Nitrogen-substituted hexahydropyrazino[1,2-a]pyrimidine-4,7-dione derivatives, method for the production and use thereof as medicaments
WO2004072076A1 (en) 2003-02-13 2004-08-26 Aventis Pharma Deutschland Gmbh Substituted hexahydropyrazino(1,2-a)pyrimidin-4,7-dion derivatives, method for the production and use thereof as medicaments
WO2004078261A1 (en) 2003-03-07 2004-09-16 The University Court Of The University Of Aberdeen Cannabinoid receptor inverse agonists and neutral antagonists as therapeutic agents for the treatment of bone disorders
WO2004108728A1 (en) 2003-06-09 2004-12-16 Pfizer Products Inc. Cannabinoid receptor ligands and uses thereof
WO2005061489A1 (en) 2003-12-24 2005-07-07 Prosidion Limited Heterocyclic derivatives as gpcr receptor agonists
WO2006067531A1 (en) 2004-12-24 2006-06-29 Prosidion Ltd G-protein coupled receptor (gpr116) agonists and use thereof for treating obesity and diabetes
WO2006067532A1 (en) 2004-12-24 2006-06-29 Prosidion Ltd G-protein coupled receptor agonists
WO2006070208A1 (en) 2004-12-31 2006-07-06 Prosidion Ltd. Pyridine, pyrimidine and pyrazine derivatives as gpcr agonists
WO2007003961A2 (en) 2005-06-30 2007-01-11 Prosidion Limited Gpcr agonists
WO2007003964A1 (en) 2005-06-30 2007-01-11 Prosidion Limited G-protein coupled receptor agonists
WO2007003962A2 (en) 2005-06-30 2007-01-11 Prosidion Limited Gpcr agonists
WO2007003960A1 (en) 2005-06-30 2007-01-11 Prosidion Limited Gpcr agonists
WO2007116229A1 (en) * 2006-04-06 2007-10-18 Prosidion Limited Heterocyclic gpcr agonists

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
J ORG. CHEM., vol. 68, 2003, pages 7316 - 7321
TETRAHEDRON, vol. 55, 1999, pages 11619 - 11639

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009034388A1 (en) 2007-09-10 2009-03-19 Prosidion Limited Compounds for the treatment of metabolic disorders
WO2010004347A1 (en) * 2008-07-10 2010-01-14 Prosidion Limited Heterocyclic gpcr agonists
WO2010004345A1 (en) * 2008-07-10 2010-01-14 Prosidion Limited Piperidinyl gpcr agonists
WO2010004348A1 (en) * 2008-07-10 2010-01-14 Prosidion Limited Heterocyclic gpcr agonists
US20110230507A1 (en) * 2008-07-10 2011-09-22 Prosidion Limited Piperidine GPCR Agonists
WO2010103333A1 (en) 2009-03-12 2010-09-16 Prosidion Limited Compounds for the treatment of metabolic disorders
WO2010103334A1 (en) 2009-03-12 2010-09-16 Prosidion Limited Compounds for the treatment of metabolic disorders
WO2010103335A1 (en) 2009-03-12 2010-09-16 Prosidion Limited Compounds for the treatment of metabolic disorders
US8293729B2 (en) 2009-06-24 2012-10-23 Boehringer Ingelheim International Gmbh Compounds, pharmaceutical composition and methods relating thereto
US8481731B2 (en) 2009-06-24 2013-07-09 Boehringer Ingelheim International Gmbh Compounds, pharmaceutical composition and methods relating thereto
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011113947A1 (en) 2010-03-18 2011-09-22 Boehringer Ingelheim International Gmbh Combination of a gpr119 agonist and the dpp-iv inhibitor linagliptin for use in the treatment of diabetes and related conditions
WO2011128394A1 (en) 2010-04-14 2011-10-20 Prosidion Limited 3-substituted 5-(pyrrolidine-1-carbonyl) pyrrolidine and its derivatives for use in the treatment of metabolic disorders
WO2011128395A1 (en) 2010-04-14 2011-10-20 Prosidion Limited N- substituted 3-amino 4 - ( pyrrolidine - 1 - carbonyl) pyrrolidine and its derivatives for use in the treatment of metabolic disorders
WO2011147951A1 (en) 2010-05-28 2011-12-01 Prosidion Limited Cycloamino derivatives as gpr119 antagonists
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2012066077A1 (en) 2010-11-18 2012-05-24 Prosidion Limited 1,4 di substituted pyrrolidine - 3 - yl -amine derivatives and their use for the treatment of metabolic disorders
WO2012101292A1 (en) * 2011-01-25 2012-08-02 Viviabiotech, S.L. 1,2,4-oxadiazole derivatives as drugs modulating the glp-1 peptide receptor
US9050347B2 (en) 2011-01-25 2015-06-09 Viviabiotech, S.L. 1,2,4-oxadiazole derivatives as drugs modulating the GLP-1 peptide receptor
GB2488360A (en) * 2011-02-25 2012-08-29 Prosidion Ltd Heterocyclic GPCR agonists
WO2012170867A1 (en) 2011-06-09 2012-12-13 Rhizen Pharmaceuticals Sa Novel compounds as modulators of gpr-119
WO2013026587A1 (en) 2011-08-22 2013-02-28 Prosidion Limited 1,4 disubstituted pyrrolidine - 3 - yl -amine derivatives and their use for the treatment of metabolic disorders
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors

Also Published As

Publication number Publication date
EP2114936A1 (en) 2009-11-11
BRPI0806312A2 (en) 2011-09-06
EA016507B1 (en) 2012-05-30
CA2674360A1 (en) 2008-07-10
US20100048625A1 (en) 2010-02-25
JP2010514832A (en) 2010-05-06
EA200900878A1 (en) 2010-02-26

Similar Documents

Publication Publication Date Title
EP2114935B1 (en) Piperidine gpcr agonists
WO2008081208A1 (en) Piperidine gpcr agonists
EP2114933B1 (en) Piperidine gpcr agonists
EP2321308B9 (en) Piperidine gpcr agonists
EP2318399B1 (en) Piperidinyl gpcr agonists
EP2114931B1 (en) Piperidine gpcr agonists
US8173807B2 (en) Pyridine, pyrimidine and pyrazine derivatives as GPCR agonists
EP2114932A1 (en) Piperidine gpcr agonists
WO2010004345A1 (en) Piperidinyl gpcr agonists
EP2010485A1 (en) Azetidine derivatives as g-protein coupled receptor (gpr119 ) agonists
WO2010004348A1 (en) Heterocyclic gpcr agonists

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880007022.3

Country of ref document: CN

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

Ref document number: 08702097

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2426/KOLNP/2009

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2674360

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2009544451

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 200900878

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 2008702097

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12522030

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0806312

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20090706