WO2008076042A1 - Azetidinpiperazine derivatives that are neurokinin (nk) receptor antagonists and their use. - Google Patents

Azetidinpiperazine derivatives that are neurokinin (nk) receptor antagonists and their use. Download PDF

Info

Publication number
WO2008076042A1
WO2008076042A1 PCT/SE2007/001115 SE2007001115W WO2008076042A1 WO 2008076042 A1 WO2008076042 A1 WO 2008076042A1 SE 2007001115 W SE2007001115 W SE 2007001115W WO 2008076042 A1 WO2008076042 A1 WO 2008076042A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
compounds
formula
methyl
compound according
Prior art date
Application number
PCT/SE2007/001115
Other languages
French (fr)
Inventor
Sara Holmqvist
Original Assignee
Astrazeneca Ab
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Astrazeneca Ab filed Critical Astrazeneca Ab
Publication of WO2008076042A1 publication Critical patent/WO2008076042A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants

Definitions

  • Azetidinpiperazine derivatives that are Neurokinin (NK) receptor antagonists and their use.
  • the present invention relates to new compounds of formula I, to pharmaceutical compositions comprising said compounds, and to the use of said compounds in therapy.
  • the present invention further relates to processes for the preparation of compounds of formula I.
  • the neurokinins also known as the tachykinins, comprise a class of peptide neurotransmitters which are found in the peripheral and central nervous systems.
  • the three principal tachykinins are Substance P (SP), Neurokinin A (NKA) and Neurokinin B
  • NKJB neurokinin 1
  • NK 2 neurokinin 2
  • NK 3 neurokinin 3
  • NK receptor antagonist for the treatment of e.g. respiratory, cardiovascular, neuro, pain, oncology, inflammatory and/or gastrointestinal disorders.
  • respiratory, cardiovascular, neuro, pain, oncology, inflammatory and/or gastrointestinal disorders In order to increase the therapeutic index of such therapy it is desirable to obtain such a compound possessing no or minimal toxicity as well as being selective to said NK receptors.
  • said medicament has favourable pharmacokinetic and metabolic properties thus providing an improved therapeutic and safety profile such as lower liver enzyme inhibiting properties.
  • the aperture-forming alpha sub-units are encoded by the human ether-a-go-go-related gene (hERG). Since IKr plays a key role in repolarisation of the cardiac action potential, its inhibition slows repolarisation and this is manifested as a prolongation of the QT interval. Whilst QT interval prolongation is not a safety concern per se, it carries a risk of cardiovascular adverse effects and in a small percentage of people it can lead to TdP and degeneration into ventricular fibrillation.
  • hERG human ether-a-go-go-related gene
  • the NK receptor antagonist has a suitable balance of pharmacodynamic and pharmacokinetic properties to make it therapeutically useful.
  • the NK receptor antagonist needs to be balanced with regard to relevant pharmacokinetic properties.
  • the NK antagonist has: a) sufficiently high affinities at the different NK receptors, b) pharmacokinetic properties (absorption, distribution and elimination properties) that makes it possible for the drug to act at the targeted NK receptors in the CNS as well as in the periphery.
  • the NK receptor antagonist needs to have sufficiently high metabolic stability, c) sufficiently low affinities to different ion channels, such as the hERG-encoded potassium channel in order to obtain a tolerable safety profile and d) liver enzyme (such as CYP3A4) inhibiting properties at a low level to prevent drug-drug interactions.
  • NK receptor antagonist Furthermore, in order to enhance the efficacy of the NK receptor antagonist, it is beneficial to have an NK antagonist with a long-lasting competitive mode of action at the receptor.
  • EP 0625509, EP 0630887, WO 95/05377, WO95/12577, WO 95/15961, WO 96/24582, WO 00/02859, WO 00/20003, WO 00/20389, WO 00/25766, WO 00/34243, WO 02/51807 and WO 03/037889 disclose piperidinylbutylamide derivatives, which are tachykinin antagonists.
  • NK2 Receptor Human Neurokinin-2 Receptor
  • Roderick MacKenzie,A., et al, Bioorganic & Medicinal Chemistry Letters (2003), 13, 2211-2215 discloses the compound N " -[2-(3,4-dichlorophenyl)-4-(3-morphoIin-4-ylazetidin-l-yl)butyl]-iV- methylbenzamide which was found to possess functional NK 2 receptor antagonistic properties.
  • WO 96/05193, WO 97/27185 and EP 0962457 disclose azetidinylalkyllactam derivatives with tachykinin antagonist activity.
  • EP 0790248 discloses azetidinylalkylazapiperidones and azetidinylalkyloxapiperidones, which are stated to be tachykinin antagonists.
  • WO 99/01451 and WO 97/25322 disclose azetidinylalkylpiperidine derivatives claimed to be tachykinin antagonists.
  • EP 0791592 discloses azetidinylalkylglutarimides with tachykinin antagonistic properties.
  • WO2004/110344 A2 discloses dual NKl, 2 antagonists and the use thereof.
  • An object of the present invention was to provide novel neurokinin antagonists useful in therapy.
  • a further object was to provide novel compounds having well-balanced pharmacokinetic and pharmacodynamic properties.
  • the present invention provides a compound of the general formula (I)
  • Ar is selected from
  • the present invention relates to compounds of formula I as defined above as well as to salts thereof.
  • Salts for use in pharmaceutical compositions will be pharmaceutically acceptable salts, but other salts may be useful in the production of the compounds of formula I.
  • the compounds of the present invention are capable of forming salts with various inorganic and organic acids and such salts are also within the scope of this invention.
  • acid addition salts include acetate, adipate, ascorbate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, citrate, cyclohexyl sulfamate, ethanesulfonate, fumarate, glutamate, glycolate, hemisulfate, 2- hydroxyethylsulfonate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, hydroxymaleate, lactate, malate, maleate, methanesulfonate, 2-naphthalenesulfonate, nitrate, oxalate, palmoate, persulfate, phenylacetate, phosphate, picrate, pivalate, propionate
  • Non-pharmaceutically-acceptable salts may be prepared from the corresponding acid in conventional manner.
  • Non-pharmaceutically-acceptable salts may be useful as intermediates and as such are another aspect of the present invention.
  • Acid addition salts may also be in the form of polymeric salts such as polymeric sulfonates.
  • the salts may be formed by conventional means, such as by reacting the free base form of the product with one or more equivalents of the appropriate acid in a solvent or medium in which the salt is poorly soluble, or in a solvent such as water, which is removed in vacuo or by freeze drying or by exchanging the anions of an existing salt for another anion on a suitable ion-exchange resin.
  • the compounds according to formula (I) can be in the form of the single stereoisomers, i.e. the single enantiomer (the i?-enantiomer or the iS'-enantiomer).
  • the compounds according to formula (I) can also be in the form of a racemic mixture, i.e. an equimolar mixture of enantiomers.
  • the compounds according to formula (I) can be in the form of a non equimolar mixture of enantiomers.
  • the compounds can exist as a mixture of conformational isomers.
  • the compounds of this invention comprise both mixtures of, and individual, conformational isomers.
  • a pharmaceutical formulation comprising a compound of formula I, as a single enantiomer, a racemate or a mixture thereof as a free base or pharmaceutically acceptable salts thereof, for use in prevention and/or treatment of respiratory, cardiovascular, neuro, pain, oncology, imflammatory and/or gastrointestinal disorders.
  • compositions of this invention may be administered in standard manner for the disease condition that it is desired to treat, for example by oral, topical, parenteral, buccal, nasal, vaginal or rectal administration or by inhalation or insufflation.
  • the compounds of this invention may be formulated by means known in the art into the form of, for example, tablets, pellets, capsules, aqueous or oily solutions, suspensions, emulsions, creams, ointments, gels, nasal sprays, suppositories, finely divided powders or aerosols or nebulisers for inhalation, and for parenteral use (including intravenous, intramuscular or infusion) sterile aqueous or oily solutions or suspensions or sterile emulsions.
  • composition of this invention may also contain, or be co-administered (simultaneously or sequentially) with, one or more pharmacological agents of value in treating one or more disease conditions referred to herein.
  • compositions of this invention will normally be administered to humans in a daily dose of a compound of formula I of from 0.01 to 25 mg/kg body weight.
  • a daily dose of the compound of formula I from 0.1 to 5 mg/kg body weight is administered.
  • This daily dose may be given in divided doses as necessary, the precise amount of the compound administered and the route of administration depending on the weight, age and sex of the patient being treated and on the particular disease condition being treated according to principles known in the art.
  • unit dosage forms will contain about 1 mg to 500 mg of a compound of this invention.
  • a tablet or capsule for oral administration may conveniently contain up to 250 mg (and typically 5 to 100 mg) of a compound of the formula (I) or a pharmaceutically acceptable salt thereof.
  • a compound of the formula (I) or a pharmaceutically acceptable salt thereof may be administered in a daily dosage range of from 5 to 100 mg, in a single dose or divided into two to four daily doses.
  • a sterile solution or suspension containing up to 10% w/w (and typically 5% w/w) of a compound of the formula (I) or a pharmaceutically acceptable salt thereof may be used.
  • the present invention provides a method of treating or preventing a disease condition wherein antagonism of tachykinins acting at the NK receptors is beneficial which comprises administering to a subject an effective amount of a compound of the formula (I) or a pharmaceutically-acceptable salt thereof.
  • the present invention also provides the use of a compound of the formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicament for use in a disease condition wherein antagonism of tachykinins acting at the NK receptors is beneficial.
  • the compounds of formula (I) or pharmaceutically acceptable salts or solvates thereof may be used in the manufacture of a medicament for use in the prevention or treatment of respiratory, cardiovascular, neuro, pain, oncology and/or gastrointestinal disorders.
  • disorders are asthma, allergic rhinitis, pulmonary diseases, cough, cold, inflammation, chronic obstructive pulmonary disease, airway reactivity, urticaria,hypertension, rheumatoid arthritis, edema, angiogenesis, pain, migraine, tension headache, psychoses, depression, anxiety, Alzheimer's disease, schizophrenia, Huntington's disease, bladder hypermotility, urinary incontinence, eating disorder, manic depression, substance dependence, movement disorder, cognitive disorder, obesity, stress disorders, micturition disorders, mania, hypomania and aggression, bipolar disorder, cancer, carcinoma, fibromyalgia, non cardiac chest pain, gastrointestinal hypermotility, gastric asthma, Crohn's disease, gastric emptying disorders, ulcerative colitis, irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), emesis, gastric asthma, gastric motility disorders, gastro-esophageal reflux disease (GERD) or functional dyspepsia.
  • IBS irritable bowel syndrome
  • the present invention provides a process for preparing a compound of the formula (I) or salts thereof which process comprises:
  • Ar is as hereinbefore defined; and L' is a leaving group; and optionally forming a pharmaceutically acceptable salt.
  • the compounds of the formulae (II) and (III) are reacted under conditions of reductive alkylation.
  • the reaction is typically performed at a non-extreme temperature, for example 0 - 40 0 C, in a substantially inert solvent for example dichloromethane.
  • Typical reducing agents include borohydrides such as sodium cyanoborohydride.
  • the compounds of the formulae (II) and (IV) are reacted under conditions of alkylation.
  • L is a leaving group such as halogen or alkylsulfonyloxy.
  • the reaction is typically performed at an elevated temperature, for example 30 - 130 0 C, in a substantially inert solvent for example DMF.
  • the compounds of the formula (II) may be prepared in conventional manner.
  • the compounds of the formula (DI) may be prepared, for example, by reacting a compound of the formula (VI) with a compound of the formula (VII):
  • the compounds of the formula (IV) may be prepared, for example, by reacting a compound of the formula (VI) with a compound of the formula (VIII):
  • acylation reaction is performed in the presence of a non-nucleophilic base, for example i ⁇ N-diisopropylethylamine, in a substantially inert solvent such as dichloromethane at a non-extreme temperature.
  • the compounds of the present invention most often show highly complex NMR spectra due to the existence of conformational isomers. This is believed to be a result from slow rotation about the amide and/or aryl bond.
  • the following abbreviations are used in the presentation of the NMR data of the compounds: s-singlet; d- doublet; t-triplet; qt-quartet; qn-quintet; m-multiplet; b-broad; cm-complex multiplet, which may include broad peaks.
  • the mixture was stirred for 2 h, diluted with water and then extracted twice with methylene chloride.
  • the combined organic solutions were separated by means of a phase separator column and the solvent was removed by evaporation.
  • the residue (1.08 g) was dissolved in THF (18 mL) and water (4.5 mL) and to the resultant solution was added NaIO 4 (0.73 g, 3.4 mmol).
  • the mixture was stirred under nitrogen overnight at RT.
  • the mixture was partitioned between methylene chloride and water.
  • the aqueous phase was extracted with methylene chloride and then the combined organic solutions were washed with brine and separated by means of a phase separator column.
  • CHO Kl cells obtained from ATCC were stably transfected with the human NK 2 receptor (hNK 2 R cDNA in pRc/CMV, Invitrogen) or the human NK 3 receptor (hNK 3 R in pcDNA 3.1/Hygro (+)/IRES/CD8, Invitrogen vector modified at AstraZeneca EST-Bio UK, Alderley Park).
  • the cells were transfected with the cationic lipid reagent LIPOFECTAMINETM (Invitrogen) and selection was performed with Geneticin (G418, Invitrogen) at lmg/ml for the hNK 2 R transfected cells and with Hygromycin (Invitrogen) at 500 ⁇ g/ml for the KNK 3 R transfected cells.
  • Single cell clones were collected by aid of Fluorescence Activated Cell Sorter (FACS), tested for functionality in a FLIPR assay (see below), expanded in culture and cryopreserved for future use.
  • CHO cells stably transfected with human NK 1 receptors originates from AstraZeneca R&D, Wilmington USA.
  • Human NK 1 receptor cDNA obtained from RNA- PCR from lung tissue was subcloned into pRcCMV (Invitrogen). Transfection was performed by Calcium Phosphate and selection with lmg/ml G418.
  • the CHO cells stably transfected with KNK 1 R, KNK 2 R and KNK 3 R were cultured in a humidified incubator under 5% CO 2 , in Nut Mix F12 (HAM) with Glutamax 1, 10% Foetal Bovine Serum (FBS), 1% Penicillin/Streptomycin (PEST) supplemented with 200 ⁇ g/ml Geneticin for the KNK 1 R and KNK 2 R expressing cells and 500 ⁇ g/ml Hygromycin for the KNK 3 R expressing cells.
  • the cells were grown in Tl 75 flasks and routinely passaged when 70-80% confluent for up to 20-25 passages.
  • NK1/NK2/NK3 receptor mediated increase in intracellular Ca 2+ was assessed by the following procedure: CHO cells stably transfected with human NK 1 , NK 2 or NK 3 receptors were plated in black walled/clear bottomed 96-well plates (Costar 3904) at 3.5x10 4 cells per well and grown for approximately 24h in normal growth media in a 37°C CO 2 -incubator. Before the FLIPR assay the cells of each 96-well plate were loaded with the Ca 2+ sensitive
  • K B IC 50 / 1+ (EC 60 cone, of agonist used in assay / EC50 agonist) .
  • pK B - log K B 5 Determining the Dissociation Constant (Ki) of compounds for Human NK X ZNK 2 ZNK S Receptors (Binding Assay)
  • Membranes were prepared from CHO cells stably transfected with human NKi, NK 2 or NK 3 receptors according to the following method.
  • Cells were detached with Accutase® solution, harvested in PBS containing 5% FBS by0 centrifugation, washed twice in PBS and resuspended to a concentration of 1x10 8 cells/ml in Tris-HCl 50 mM, KCl 300 mM, EDTA-N 2 10 mM pH 7.4 (4°C).
  • Cell suspensions were homogenized with an UltraTurrax 30 s 12.000 rpm. The homogenates were centrifuged at 38.000 x g (4 0 C) and the pellet resuspended in Tris-HCl 50 mM pH 7.4. The homogenization was repeated once and the homogenates were incubated on ice for 45 min.
  • the radioligand binding assay is performed at room temperature in 96-well microtiter plates (No-binding Surface Plates, Corning 3600) with a final assay volume of 200 ⁇ l/well in incubation buffer (5OmM Tris buffer (pH 7.4 RT) containing 0.1 % BSA, 40 mg/L Bacitracin, complete EDTA-free protease inhibitor cocktail tablets 20 pills/L (Roche) and 3mM MnCl 2 ).
  • Competition binding curves were done by adding increasing amounts of the test compound. Test compounds were dissolved and serially diluted in DMSO, final
  • radioligand 50 ⁇ l radioligand, 3 ⁇ l test compound diluted in DMSO and 47 ⁇ l incubation buffer were mixed with 5-10 ⁇ g cell membranes in lOO ⁇ l incubation buffer and incubated for 30 min at room temperature on a microplate shaker. The membranes were then collected by rapid filtration on Filtermat B(Wallac), presoaked in 0.1% BSA and 0.3% Polyethyleneimine (Sigma P-3143), using a Micro 96 Harvester (Skatron Instruments, Norway). Filters were washed by the harvester with ice-cold wash buffer (5OmM Tris-HCl, pH 7.4 at 4°C, containing 3mM MnCl 2 ) and dried at 5O 0 C for 30- 60 min.
  • ice-cold wash buffer 5OmM Tris-HCl, pH 7.4 at 4°C, containing 3mM MnCl 2
  • the compounds of the invention demonstrated statistically significant antagonistic activity at the NKi receptor within the range of 8-9 for the pKg .
  • the range for the pK ⁇ was 7-8.
  • the antagonistic activity at the NK3 receptor was 8-9 for the pKg.
  • the compounds of the invention demonstrated statistically significant CYP3 A4 inhibition at a low level.
  • the IC50 values tested according to Bapiro et al; Drug Metab. Dispos. 29, 30-35 (2001) were generally greater than 30 ⁇ M.
  • the compounds of the invention which were tested, demonstrated statistically significant hERG activity at a low level.
  • the IC50 values tested as described above were generally greater than 5 ⁇ M.
  • the rate of biotransformation can be measured as either metabolite(s) formation or the rate of disappearance of the parent compound.
  • the experimental design involves incubation of low concentrations of substrate (usually 1.0 ⁇ M) with liver microsomes (usually 0.5 mg/ml) and taking out aliquotes at varying time points (usually 0, 5, 10, 15, 20, 30, 40 min.).
  • the test compound is usually dissolved in DMSO.
  • 10 incubation mixture is usually 0.1% or less since more solvent can drastically reduce the activities of some CYP450s. Incubations are done in 100 mM potassium phosphate buffer, pH 7.4 and at 37 °C. Acetonitrile or methanol is used to stop the reaction. The parent compound is analysed by HPLC-MS. From the calculated half-life, t ⁇ a, the intrinsic clearance, Clint, is estimated by taking microsomal protein concentration and liver weight
  • the compounds of the invention had in vitro metabolic stability at a high level. Intrinsic clearance values tested as above were generally lower than 120 ⁇ l/min/mg protein.
  • Male Mongolian gerbils (60-8Og) are purchased from Charles River, Germany. On arrival, they are housed in groups often, with food and water ad libitum in temperature and humidity-controlled holding rooms. The animals are allowed at least 7 days to acclimatize to the housing conditions before experiments. Each animal is used only once and euthanized immediately after the experiment by heart punctuation or a lethal overdose of pentobarbital sodium.
  • Gerbils are anaesthetized with isoflurane.
  • Potential CNS-permeable NKl receptor antagonists are administered intraperitoneally, intravenously or subcutaneously. The compounds are given at various time points (typically 30-120 minutes) prior to stimulation with agonist.
  • the gerbils are lightly anaesthetized using isofluorane and a small incision is made in the skin over bregma.
  • 10 pmol of ASMSP a selective NKl receptor agonist
  • ASMSP a selective NKl receptor agonist
  • NKl receptor antagonists at peripheral NKl receptors can be assessed in gerbils in vivo using the so-called chrornodacryorrhea model (Bristow LJ, Young L. Chrornodacryorrhea and repetitive hind paw tapping: models of peripheral and central tachykinin NKl receptor 5 activation in gerbils. Eur J Pharmacol 1994; 253: 245-252). Briefly, systemic (intravenous) administration of NKl receptor agonists to anaesthetized gerbils results in profuse secretion of red/brown tears in the eyes due to porphyrin secretion from the Harderian gland. NKl receptor antagonists block NKl -agonist evoked chromodacryorrhea. o Fecal pellet output (NK2 specific test model)
  • NK2 receptor agonist-induced fecal pellet output can be determined by measuring NK2 receptor agonist-induced fecal pellet output using gerbil as described in e.g. The Journal of Pharmacology and Experimental Therapeutics (2001), pp. 559-564. 5
  • Colorectal distension (CPJD) in gerbils is performed as previously described in rats and mice (Tammpere A, Brusberg M, Axenborg J, Hirsch I, Larsson H, Lindstr ⁇ m E. Evaluation of pseudo-affective responses to noxious colorectal distension in rats byo manometric recordings. Pain 2005; 116: 220-226; Arvidsson S, Larsson M, Larsson H, Lindstr ⁇ m E, Martinez V. Assessment of visceral pain-related pseudo-affective responses to colorectal distension in mice by intracolonic manometric recordings. J Pain 2006; 7: 108-118) with slight modifications.
  • gerbils are habituated to Bollmann cages 30- 60 min per day for three consecutive days prior to experiments to reduce motion artefactss due to restraint stress.
  • a 2 cm polyethylene balloon (made in-house) with connecting catheter is inserted in the distal colon, 2 cm from the base of the balloon to the anus, during light isoflurane anaesthesia (Forene ® , Abbott Scandinavia AB, Solna, Sweden).
  • the catheter is fixed to the tail with tape.
  • the balloons are connected to pressure transducers (P-602, CFM-k33, 100 mmHg, Bronkhorst HI-TEC, Veenendal, The Netherlands).
  • Gerbils are allowed to recover from sedation in the Bollmann cages for at least 15 min before- the start of experiments.
  • a customized barostat (AstraZeneca, M ⁇ lndal, Sweden) is used to manage air inflation and balloon pressure control.
  • a customized computer software (PharmLab on-line 4.0) running on a standard computer is used to control the barostat and to perform data collection.
  • the distension paradigm used consists of 12 repeated phasic distensions at 80 mmHg, with a pulse duration of 30 sec at 5 min intervals.
  • Compounds or their respective vehicle are administered as intraperitoneal (i.p.) injections before the CRD paradigm.
  • Each gerbil receives both vehicle and compound on different occasions with at least two days between experiments. Hence, each gerbil serves as its own vehicle control.
  • the analog input channels are sampled with individual sampling rates, and digital filtering is performed on the signals.
  • the balloon pressure signals are sampled at 50 samples/s.
  • a highpass filter at 1 Hz is used to separate the contraction-induced pressure changes from the slow varying pressure generated by the barostat.
  • a resistance in the airflow between the pressure generator and the pressure transducer further enhances the pressure variations induced by abdominal contractions of the animal.
  • a customized computer software (PharmLab off-line 4.0) is used to quantify the magnitude of highpass-filtered balloon pressure signals.
  • the average rectified value (ARV) of the highpass-filtered balloon pressure signals is calculated for 30 s before the pulse (i.e baseline reponse) and for the duration of the pulse.

Landscapes

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

Abstract

The present invention relates to new compounds of formula (I), to pharmaceutical compositions comprising said compounds, and to the use of said compounds in therapy. The present invention further relates to processes for the preparation of compounds of formula (I).

Description

Azetidinpiperazine derivatives that are Neurokinin (NK) receptor antagonists and their use.
Field of the Invention
The present invention relates to new compounds of formula I, to pharmaceutical compositions comprising said compounds, and to the use of said compounds in therapy. The present invention further relates to processes for the preparation of compounds of formula I.
Background of the invention
The neurokinins, also known as the tachykinins, comprise a class of peptide neurotransmitters which are found in the peripheral and central nervous systems. The three principal tachykinins are Substance P (SP), Neurokinin A (NKA) and Neurokinin B
(NKJB). At least three receptor types are known for the three principal tachykinins. Based upon their relative selectivities favouring the agonists SP, NKA and NKB, the receptors are classified as neurokinin 1 (NK1), neurokinin 2 (NK2) and neurokinin 3 (NK3) receptors, respectively.
There is a need for an orally active NK receptor antagonist for the treatment of e.g. respiratory, cardiovascular, neuro, pain, oncology, inflammatory and/or gastrointestinal disorders. In order to increase the therapeutic index of such therapy it is desirable to obtain such a compound possessing no or minimal toxicity as well as being selective to said NK receptors. Furthermore, it is considered necessary that said medicament has favourable pharmacokinetic and metabolic properties thus providing an improved therapeutic and safety profile such as lower liver enzyme inhibiting properties.
It is well known that certain compounds may cause undesirable effects on cardiac repolarisation in man, observed as a prolongation of the QT interval on electrocardiograms (ECG). In extreme circumstances, this drug-induced prolongation of the QT interval can lead to a type of cardiac arrhythmia called Torsades de Pointes (TdP; Vandenberg et al. hERG K+ channels: friend and foe. Trends Pharmacol Sci 2001; 22: 240-246), leading ultimately to ventricular fibrillation and sudden death. The primary event in this syndrome is inhibition of the rapid component of the delayed rectifying potassium current (IKr) by these compounds. The compounds bind to the aperture-forming alpha sub-units of the channel protein carrying this current. The aperture-forming alpha sub-units are encoded by the human ether-a-go-go-related gene (hERG). Since IKr plays a key role in repolarisation of the cardiac action potential, its inhibition slows repolarisation and this is manifested as a prolongation of the QT interval. Whilst QT interval prolongation is not a safety concern per se, it carries a risk of cardiovascular adverse effects and in a small percentage of people it can lead to TdP and degeneration into ventricular fibrillation.
In particular, it is desirable that the NK receptor antagonist has a suitable balance of pharmacodynamic and pharmacokinetic properties to make it therapeutically useful. In addition to having sufficient and selective potency, the NK receptor antagonist needs to be balanced with regard to relevant pharmacokinetic properties. Thus, it is necessary that the NK antagonist has: a) sufficiently high affinities at the different NK receptors, b) pharmacokinetic properties (absorption, distribution and elimination properties) that makes it possible for the drug to act at the targeted NK receptors in the CNS as well as in the periphery. For instance, the NK receptor antagonist needs to have sufficiently high metabolic stability, c) sufficiently low affinities to different ion channels, such as the hERG-encoded potassium channel in order to obtain a tolerable safety profile and d) liver enzyme (such as CYP3A4) inhibiting properties at a low level to prevent drug-drug interactions.
Furthermore, in order to enhance the efficacy of the NK receptor antagonist, it is beneficial to have an NK antagonist with a long-lasting competitive mode of action at the receptor.
EP 0625509, EP 0630887, WO 95/05377, WO95/12577, WO 95/15961, WO 96/24582, WO 00/02859, WO 00/20003, WO 00/20389, WO 00/25766, WO 00/34243, WO 02/51807 and WO 03/037889 disclose piperidinylbutylamide derivatives, which are tachykinin antagonists.
"4-Amino-2-(aryl)-butylbenzamides and Their Conformationally Constrained Analogues. Potent Antagonists of the Human Neurokinin-2 (NK2) Receptor", Roderick MacKenzie,A., et al, Bioorganic & Medicinal Chemistry Letters (2003), 13, 2211-2215, discloses the compound N"-[2-(3,4-dichlorophenyl)-4-(3-morphoIin-4-ylazetidin-l-yl)butyl]-iV- methylbenzamide which was found to possess functional NK2 receptor antagonistic properties.
WO 96/05193, WO 97/27185 and EP 0962457 disclose azetidinylalkyllactam derivatives with tachykinin antagonist activity.
EP 0790248 discloses azetidinylalkylazapiperidones and azetidinylalkyloxapiperidones, which are stated to be tachykinin antagonists.
WO 99/01451 and WO 97/25322 disclose azetidinylalkylpiperidine derivatives claimed to be tachykinin antagonists.
EP 0791592 discloses azetidinylalkylglutarimides with tachykinin antagonistic properties.
WO2004/110344 A2 discloses dual NKl, 2 antagonists and the use thereof.
An object of the present invention was to provide novel neurokinin antagonists useful in therapy. A further object was to provide novel compounds having well-balanced pharmacokinetic and pharmacodynamic properties. Outline of the invention
The present invention provides a compound of the general formula (I)
Figure imgf000005_0001
(I) wherein
Ar is selected from
Figure imgf000005_0002
as well as pharmaceutically and pharmacologically acceptable salts thereof, and enantiomers of the compound of formula I and salts thereof.
The present invention relates to compounds of formula I as defined above as well as to salts thereof. Salts for use in pharmaceutical compositions will be pharmaceutically acceptable salts, but other salts may be useful in the production of the compounds of formula I.
The compounds of the present invention are capable of forming salts with various inorganic and organic acids and such salts are also within the scope of this invention. Examples of such acid addition salts include acetate, adipate, ascorbate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, citrate, cyclohexyl sulfamate, ethanesulfonate, fumarate, glutamate, glycolate, hemisulfate, 2- hydroxyethylsulfonate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, hydroxymaleate, lactate, malate, maleate, methanesulfonate, 2-naphthalenesulfonate, nitrate, oxalate, palmoate, persulfate, phenylacetate, phosphate, picrate, pivalate, propionate, quinate, salicylate, stearate, succinate, sulfamate, sulfanilate, sulfate, tartrate, tosylate (p-toluenesulfonate), and undecanoate.
Pharmaceutically acceptable salts may be prepared from the corresponding acid in conventional manner. Non-pharmaceutically-acceptable salts may be useful as intermediates and as such are another aspect of the present invention.
Acid addition salts may also be in the form of polymeric salts such as polymeric sulfonates.
The salts may be formed by conventional means, such as by reacting the free base form of the product with one or more equivalents of the appropriate acid in a solvent or medium in which the salt is poorly soluble, or in a solvent such as water, which is removed in vacuo or by freeze drying or by exchanging the anions of an existing salt for another anion on a suitable ion-exchange resin.
Compounds of formula I have one chiral centre, and it is to be understood that the invention encompasses all stereoisomers. The compounds according to formula (I) can be in the form of the single stereoisomers, i.e. the single enantiomer (the i?-enantiomer or the iS'-enantiomer). The compounds according to formula (I) can also be in the form of a racemic mixture, i.e. an equimolar mixture of enantiomers. Furthermore, the compounds according to formula (I) can be in the form of a non equimolar mixture of enantiomers.
The compounds can exist as a mixture of conformational isomers. The compounds of this invention comprise both mixtures of, and individual, conformational isomers. Pharmaceutical formulations
According to one aspect of the present invention there is provided a pharmaceutical formulation comprising a compound of formula I, as a single enantiomer, a racemate or a mixture thereof as a free base or pharmaceutically acceptable salts thereof, for use in prevention and/or treatment of respiratory, cardiovascular, neuro, pain, oncology, imflammatory and/or gastrointestinal disorders.
The pharmaceutical compositions of this invention may be administered in standard manner for the disease condition that it is desired to treat, for example by oral, topical, parenteral, buccal, nasal, vaginal or rectal administration or by inhalation or insufflation.
For these purposes the compounds of this invention may be formulated by means known in the art into the form of, for example, tablets, pellets, capsules, aqueous or oily solutions, suspensions, emulsions, creams, ointments, gels, nasal sprays, suppositories, finely divided powders or aerosols or nebulisers for inhalation, and for parenteral use (including intravenous, intramuscular or infusion) sterile aqueous or oily solutions or suspensions or sterile emulsions.
In addition to the compounds of the present invention the pharmaceutical composition of this invention may also contain, or be co-administered (simultaneously or sequentially) with, one or more pharmacological agents of value in treating one or more disease conditions referred to herein.
The pharmaceutical compositions of this invention will normally be administered to humans in a daily dose of a compound of formula I of from 0.01 to 25 mg/kg body weight. Alternatively, a daily dose of the compound of formula I from 0.1 to 5 mg/kg body weight is administered. This daily dose may be given in divided doses as necessary, the precise amount of the compound administered and the route of administration depending on the weight, age and sex of the patient being treated and on the particular disease condition being treated according to principles known in the art. Typically unit dosage forms will contain about 1 mg to 500 mg of a compound of this invention. For example a tablet or capsule for oral administration may conveniently contain up to 250 mg (and typically 5 to 100 mg) of a compound of the formula (I) or a pharmaceutically acceptable salt thereof. In another example, for administration by inhalation, a compound of the formula (I) or a pharmaceutically acceptable salt thereof may be administered in a daily dosage range of from 5 to 100 mg, in a single dose or divided into two to four daily doses. In a further example, for administration by intravenous or intramuscular injection or infusion, a sterile solution or suspension containing up to 10% w/w (and typically 5% w/w) of a compound of the formula (I) or a pharmaceutically acceptable salt thereof may be used.
Medical and pharmaceutical use
The present invention provides a method of treating or preventing a disease condition wherein antagonism of tachykinins acting at the NK receptors is beneficial which comprises administering to a subject an effective amount of a compound of the formula (I) or a pharmaceutically-acceptable salt thereof. The present invention also provides the use of a compound of the formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicament for use in a disease condition wherein antagonism of tachykinins acting at the NK receptors is beneficial.
The compounds of formula (I) or pharmaceutically acceptable salts or solvates thereof may be used in the manufacture of a medicament for use in the prevention or treatment of respiratory, cardiovascular, neuro, pain, oncology and/or gastrointestinal disorders.
Examples of such disorders are asthma, allergic rhinitis, pulmonary diseases, cough, cold, inflammation, chronic obstructive pulmonary disease, airway reactivity, urticaria,hypertension, rheumatoid arthritis, edema, angiogenesis, pain, migraine, tension headache, psychoses, depression, anxiety, Alzheimer's disease, schizophrenia, Huntington's disease, bladder hypermotility, urinary incontinence, eating disorder, manic depression, substance dependence, movement disorder, cognitive disorder, obesity, stress disorders, micturition disorders, mania, hypomania and aggression, bipolar disorder, cancer, carcinoma, fibromyalgia, non cardiac chest pain, gastrointestinal hypermotility, gastric asthma, Crohn's disease, gastric emptying disorders, ulcerative colitis, irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), emesis, gastric asthma, gastric motility disorders, gastro-esophageal reflux disease (GERD) or functional dyspepsia. Methods of preparation
In another aspect the present invention provides a process for preparing a compound of the formula (I) or salts thereof which process comprises:
a) reacting a compound of the formula (II) with a compound of the formula (III):
Figure imgf000010_0001
wherein Ar is as hereinbefore defined; and the conditions are such that reductive alkylation of the compounds of the formula (II) forms an N-C bond between the nitrogen atom of the azetidine group of the compounds of formula (II) and the carbon atom of the aldehyde group of the compounds of formula (III); or b) reacting a compound of the formula (II) with a compound of the formula (IV):
Figure imgf000011_0001
wherein Ar is as hereinbefore defined; and L is a group such that alkylation of the compounds of the formula (II) forms an N-C bond between the nitrogen atom of the azetidine group of the compounds of formula (II) and the carbon atom of the compounds of formula (IV) that is adjacent to the L group; or c) reacting a compound of the formula (V) with a compound of the formula (VI):
Figure imgf000011_0002
wherein Ar is as hereinbefore defined; and L' is a leaving group; and optionally forming a pharmaceutically acceptable salt.
The compounds of the formulae (II) and (III) are reacted under conditions of reductive alkylation. The reaction is typically performed at a non-extreme temperature, for example 0 - 40 0C, in a substantially inert solvent for example dichloromethane. Typical reducing agents include borohydrides such as sodium cyanoborohydride.
The compounds of the formulae (II) and (IV) are reacted under conditions of alkylation. Typically in the compounds of the formula (IV) L is a leaving group such as halogen or alkylsulfonyloxy. The reaction is typically performed at an elevated temperature, for example 30 - 130 0C, in a substantially inert solvent for example DMF.
The compounds of the formula (II) may be prepared in conventional manner. The compounds of the formula (DI) may be prepared, for example, by reacting a compound of the formula (VI) with a compound of the formula (VII):
Figure imgf000012_0001
(VII)
under conventional acylation conditions.
The compounds of the formula (IV) may be prepared, for example, by reacting a compound of the formula (VI) with a compound of the formula (VIII):
Figure imgf000012_0002
(VIII) wherein L is as hereinbefore defined under conventional acylation conditions. The compounds of the formulae (V) and (VT) may be reacted under conventional acylation conditions wherein
Figure imgf000013_0001
is an acid or an activated acid derivative. Such activated acid derivatives are well known in the literature. They may be formed in situ from the acid or they may be prepared, isolated and subsequently reacted. Typically I/ is chloro thereby forming the acid chloride. Typically the acylation reaction is performed in the presence of a non-nucleophilic base, for example i\ζN-diisopropylethylamine, in a substantially inert solvent such as dichloromethane at a non-extreme temperature.
The compounds of the formula (VII) and (VIII) are known or may be prepared in conventional manner.
Examples
It should be emphasised that the compounds of the present invention most often show highly complex NMR spectra due to the existence of conformational isomers. This is believed to be a result from slow rotation about the amide and/or aryl bond. The following abbreviations are used in the presentation of the NMR data of the compounds: s-singlet; d- doublet; t-triplet; qt-quartet; qn-quintet; m-multiplet; b-broad; cm-complex multiplet, which may include broad peaks.
The following example will describe, but not limit, the invention.
The following abbreviations are used in the experimental: DIPEA (N,N- diisopropylethylamine), THF (tetrahydrofuran), TBTU (ΛζΛζiV'.iV'-tetramethyl-O- (benzotriazol-1 -yl)uronium tetrafluoroborate)and RT (room temperature). Example 1
Methyl 4- π-r(3^-4-{r3-bromo-5-rtrifIuoromethyl)benzoyl¥methyl)amino|-3-f4- fluorophenyllbutyliazetidin-S-yl'l-piperazine-l-carboxylate
Figure imgf000014_0001
Methyl 4-azetidin-3-ylpiperazine-l-carboxylate (see Method 1; 160 mg, 0.080 mmol) and
3-bromo-N-[(25)-2-(4-fluorophenyl)-4-oxobutyl]-N-methyl-5-(trifluoromethyl)-benzamide (see Method 2; 54 mg, 0.12 mmol) were dissolved in methanol (15 mL) and to the resultant solution was added a mixture of zinc chloride (47 mg, 0.34 mmol), sodium cyano borohydride (38 mg, 0.60 mmol) and methanol (2 mL). The reaction mixture was stirred at RT for 15 min and then the solvent was removed by evaporation. The residue was partitioned between ethyl acetate and saturated aqueous NaHCO3. The two layers were separated and the aqueous solution was extracted twice with ethyl acetate. The organic solution was dried over Na2SO4 and then the solvent was removed by evaporation. The product was purified by means of reversed phase chromatography using a mixture of acetonitrile and aqueous 0.1 M ammonium acetate. The proper fraction were combined and concentrated on a rotavapor. To the residue was added a saturated solution OfNaHCO3. The mixture was extracted three times with ethyl acetate and the combined organic solutions were evaporated. The residue was dissolved in a mixture of acetonitrile and water and then the solvent was removed by freeze-drying. There was obtained 35 mg (69%) of the title compound as a solid. 1H NMR (500 MHz, CD3OD): 1.5-3.9 (cm, 26H), 6.9-7.5 (cm, 6H)37.9 (d, IH): m/z 630 (M+l)+. Example 2
Methyl 4-{l-r(36r)-4-{r3-chloro-5-rtrifluoromethyl)benzovn('methvnamino>-3-r4- fluorophenyDbutyllazetidin-S-yllpiperazine-l-carboxylate
Figure imgf000015_0001
Methyl 4-azetidin-3-ylpiperazine-l-carboxylate hydrochloride (see Method 1; 0.72 g, 3.1 mmol), 3-bromo-N-[(2iS)-2-(4-fluorophenyl)-4-oxobutyl]-iV-rnethyl-5-(trifluorornethyl)- benzamide (see Method 2; 0.95 g, 2.4 mmol) and triethylamine (0.66 mL, 4.7 mmol) were added to methanol (10 mL) and the mixture was stirred at RT for 45 min. Sodium triacetoxyborohydride (0.75 g, 3.5 mmol) was added in small portions during a time of 30 min. The reaction mixture was then stirred at RT for 45 min and the solvent was removed by evaporation. The residue was partitioned between methylene chloride and saturated aqueous NaHCO3. The two layers were separated by means of a phase separator column and the organic solution was evaporated. The product was purified by column chromatography on silica gel eluting with methylene chloride and ammonia saturated methanol (gradient, 98:2 - 80:20). There was obtained 0.93 g (52%) of the title compound. 1H NMR (500 MHz, CDCl3): 1.3-3.8 (cm, 26H), 6.7-7.2 (cm, 6H), 7.5 (s, IH): m/z 585 (M-I-I)+.
Preparation of Starting Materials The starting materials for the examples above are either commercially available or are readily prepared by standard methods from known materials. For example, the following reactions are an illustration, but not a limitation, of some of the starting materials. Method 1
Methyl 4-azetidin-3-ylpiperazine- 1 -carboxylate
Figure imgf000016_0001
(a) l-[l-(Diphenylmethyl)azetidin-3-yl]piperazine A mixture of l-(diphenylmethyl)azetidin-3-yl methanesulfonate (see J. Org. Chem.; 56; 1991; 6729; 25 g, 78.6 mmol), piperazine (67.7 g, 0.79 mol) and dry acetonitrile was stirred at 60°C overnight under nitrogen. The mixture was cooled and partitioned between water and methylene chloride. The organic layer was washed with water and brine. The solution was dried over Na2SO4 and then the solvent was removed by evaporation. The residue was purified by column chromatography on silica gel (methanol - methylene chloride 5:95). There was obtained 17.5 g (72%) of l-[l-(diphenylmethyl)azetidin-3- yl]piperazine as a yellow oil. 1H NMR (400 MHz, CDCl3): 2.1-2.4 (m, 4H), 2.8-2.9 (m, 2H), 3.0 (m, 4H), 3.4-3.5 (m, 2H), 3.7-3.9 (m, IH), 4.4 (s, IH), 7.2-7.4 (m, 10H); LCMS: m/z 308 (M+l)+.
(b) Methyl 4-[l-(diphenylmethyl)azetidin-3-yl]piperazine-l-carboxylate l-[l-(Diphenylmethyl)azetidin-3-yl]piperazine (0.50 g, 1.60 mmol) and N1N'- carbonyldiimidazole (0.27 g, 1.65 mmol) were dissolved in THF (5 mL). The mixture was stirred at RT for 2 h and then a solution of ammonium hydroxide (cone. 10 mL) in excess of methanol was added. The mixture was stirred at RT for two days and then excess of gaseous ammonia was introduced while cooling the mixture. The mixture was stirred at RT for 24 h and then the solvent was removed by evaporation. The residue was partitioned between ethyl acetate and saturated aqueous NaHCO3. The two layers were separated and the organic layer was dried by means of a phase separator column. The solvent was removed by evaporation And the product was purified by column chromatography on silica gel eluting with methylene chloride and methanol (gradient, 99:1 - 98:1). There was obtained 0.25 g (42%) of methyl 4-[l-(diphenylmethyl)azetidin-3-yl]piperazine-l- carboxylate. 1H NMR (500 MHz, CDCl3): 2.2-2.3 (b, 4H), 2.9 (t, 2H), 3.0 (qn, IH), 3.4 (t, 2H), 3.5 (b, 4H)73.7 (s, 3H), 4.4 (s, IH), 7.2 (t, 2H), 7.3 (t, 4H), 7.4 (d, 4H): m/z 366 (M+l)+. (c) Methyl ^azetidinS-ylpiperazine-l-carboxylate
Methyl 4-[l-(diphenylmethyl)azetidin-3-yl]piperazine-l-carboxylate (5.0 g, 13.7 mmol) was dissolved in methylene chloride, the resultant solution was cooled to 0°C, and then 1- chloroethyl chloroformate (3.9 g, 27.4 mmol) was added over a period of 5 min. The mixture was stirred at RT for 90 min and then methanol was added. The mixture was heated to reflux for 30 min and then the solvent was removed by evaporation. To the residue was added 2-prσpanol (100 mL) and the mixture was heated to reflux and then left at RT for 14 h. The product was isolated by filtration and there was obtained 3.5 g (100%) of the title compound as white powder. 1H NMR (500 MHz, CD3OD): 3.5 (b, 4H), 3.8 (s, 3H), 3.8-3.9 (b, 4H), 4.4 (m, IH), 4.5-4.6 (m, 4H).
Method 2
3-Bromo-iV-rf2.y)-2-(4-fluorophenyl)-4-oxobutyl1-JV-methyl-5-("trifluoromethyl)benzamide
Figure imgf000017_0001
(a) 3-Bromo-N-[(2S)-2-(4-fluorophenyl)pent-4-en-l-yl]-N-methyl-5-
(trifluoromethyl)benzamide
To a solution of [(2S)-2-(4-fluorophenyl)pent-4-en-l-yl]methylamine (seeBioorg. Med. Chem. Lett; 2001; 265-270; 0.54 g, 2.8 mmol) and 3-bromo-5-trifluoromethyl benzoic acid ( 0.81 g, 3.0 mmol) in DMF (7 mL) were added TBTU (0.96 g, 3.0 mmol) and DIPEA (1.41 g, 10.9 mmol). The reaction mixture was stirred under nitrogen overnight at RT and then partitioned between ethyl acetate and an aqueous NaHCO3 solution. The aqueous phase was extracted trice with ethyl acetate. The combined organic solutions were washed trice with water and then dried by a phase separator column. The solvent was removed by evaporation and the product was purified by chromatography on silica gel (ethyl acetate - heptane 10% to 17%). There was obtained 0.86 g (68%) of 3-bromo-iV-[(2JS)-2-(4- fluorophenyl)pent-4-en-l-yl]-N-methyl-5-(trifluoromethyl)benzamide. 1H ΝMR (500 MHz, CDCl3): 2.1-3.8 (cm, HH), 4.9-5.1 (m, 2H), 5.5-5.8 (m, IH), 6.8-7.4 (cm, 6H), 7.8 (s, IH).
(b) 3-Bromo-N-[(2S)-2-(4-fluorophenyl)-4-oxobutyl]-N-methyl-5- (trifluoromethyl)benzamide
To a solution of 3-bromo-N"-[(2»S)-2-(4-fluorophenyl)pent-4-en-l-yl]-iV-methyl-5- (trifluoromethyl)benzamide (0.86 g, 1.9 mmol) in acetone (45 mL) were added OsO4 (2.5% in f-butyl alcohol, 0.49 mL, 0.039 mmol) and 4-methylmorpholine-4-oxide (0.41 g, 3.5 mmol). The solution was stirred under nitrogen at RT overnight and then an aqueous solution OfNaHSO3 (39%, 45 mL) was added. The mixture was stirred for 2 h, diluted with water and then extracted twice with methylene chloride. The combined organic solutions were separated by means of a phase separator column and the solvent was removed by evaporation. The residue (1.08 g) was dissolved in THF (18 mL) and water (4.5 mL) and to the resultant solution was added NaIO4 (0.73 g, 3.4 mmol). The mixture was stirred under nitrogen overnight at RT. The mixture was partitioned between methylene chloride and water. The aqueous phase was extracted with methylene chloride and then the combined organic solutions were washed with brine and separated by means of a phase separator column. The solvent was removed by evaporation and the product was purified by chromatography on silica gel (ethyl acetate — hexane 0% to 50%). There was obtained 0.78 g (90%) of the title compound. 1H NMR (500 MHz, CDCl3): 2.4-4.4 (cm, HH), 6.2-8.2 (cm, 9H), 9.8 (s, IH); LCMS: m/z 447 (M-I)+.
Pharmacology
Transfection and culturing of cells used in FLIPR and Binding assays
Chinese Hamster Ovary (CHO) Kl cells (obtained from ATCC) were stably transfected with the human NK2 receptor (hNK2R cDNA in pRc/CMV, Invitrogen) or the human NK3 receptor (hNK3R in pcDNA 3.1/Hygro (+)/IRES/CD8, Invitrogen vector modified at AstraZeneca EST-Bio UK, Alderley Park). The cells were transfected with the cationic lipid reagent LIPOFECTAMINE™ (Invitrogen) and selection was performed with Geneticin (G418, Invitrogen) at lmg/ml for the hNK2R transfected cells and with Hygromycin (Invitrogen) at 500μg/ml for the KNK3R transfected cells. Single cell clones were collected by aid of Fluorescence Activated Cell Sorter (FACS), tested for functionality in a FLIPR assay (see below), expanded in culture and cryopreserved for future use. CHO cells stably transfected with human NK1 receptors originates from AstraZeneca R&D, Wilmington USA. Human NK1 receptor cDNA (obtained from RNA- PCR from lung tissue) was subcloned into pRcCMV (Invitrogen). Transfection was performed by Calcium Phosphate and selection with lmg/ml G418.
The CHO cells stably transfected with KNK1R, KNK2R and KNK3R were cultured in a humidified incubator under 5% CO2, in Nut Mix F12 (HAM) with Glutamax 1, 10% Foetal Bovine Serum (FBS), 1% Penicillin/Streptomycin (PEST) supplemented with 200μg/ml Geneticin for the KNK1R and KNK2R expressing cells and 500μg/ml Hygromycin for the KNK3R expressing cells. The cells were grown in Tl 75 flasks and routinely passaged when 70-80% confluent for up to 20-25 passages.
Assessing the Activity of Selected test Compounds to Inhibit Human NK1/NK2/NK3 Receptor Activation (FLIPR assay)
The activity of a compound of the invention to inhibit NKi/NKa/NKs receptor activation measured as NK1/NK2/NK3 receptor mediated increase in intracellular Ca2+ was assessed by the following procedure: CHO cells stably transfected with human NK1, NK2 or NK3 receptors were plated in black walled/clear bottomed 96-well plates (Costar 3904) at 3.5x104 cells per well and grown for approximately 24h in normal growth media in a 37°C CO2-incubator. Before the FLIPR assay the cells of each 96-well plate were loaded with the Ca2+ sensitive
5 dye Fhαo-3 (TEFLABS 0116) at 4μM in a loading media consisting of Nut Mix F12 (HAM) with Glutamax I, 22mM HEPES, 2.5mM Probenicid (Sigma P-8761) and 0.04% Pluronic F-127 (Sigma P-2443) for 1 h kept dark in a 37°C CO2-incubator. The cells were then washed three times in assay buffer (Hanks balanced salt solution (HBSS) containing 2OmM HEPES, 2.5mM Probenicid and 0.1% BSA) using a multi-channel pipette leavingo them in 150μl at the end of the last wash. Serial dilutions of a test compound in assay buffer (final DMSO concentration kept below 1%) were automatically pipetted by FLIPR (Fluorometric Imaging Plate Reader) into each test well and the fluorescence intensity was recorded (excitation 488 ran and emission 530 nm) by the FLIPR CCD camera for a 2 min pre-incubation period. 50μl of the Substance P (NK1 specific), NKA (NK2 specific), or5 Pro-7-NKB (NK3 specific) agonist solution (final concentration equivalent to an approximate EC60 concentration) was then added by FLIPR into each well already containing 200μl assay buffer (containing the test compound or vehicle) and the fluorescence was continuously monitored for another 2 min. The response was measured as the peak relative fluorescence after agonist addition and IC50S were calculated from ten-o point concentration-response curves for each compound. The IC50S were then converted to pKβ values with the following formula:
KB = IC50 / 1+ (EC60 cone, of agonist used in assay / EC50 agonist) . pKB = - log KB 5 Determining the Dissociation Constant (Ki) of compounds for Human NKXZNK2ZNKS Receptors (Binding Assay)
Membranes were prepared from CHO cells stably transfected with human NKi, NK2 or NK3 receptors according to the following method.
Cells were detached with Accutase® solution, harvested in PBS containing 5% FBS by0 centrifugation, washed twice in PBS and resuspended to a concentration of 1x108 cells/ml in Tris-HCl 50 mM, KCl 300 mM, EDTA-N210 mM pH 7.4 (4°C). Cell suspensions were homogenized with an UltraTurrax 30 s 12.000 rpm. The homogenates were centrifuged at 38.000 x g (40C) and the pellet resuspended in Tris-HCl 50 mM pH 7.4. The homogenization was repeated once and the homogenates were incubated on ice for 45 min. The homogenates were again centrifuged as described above and resuspended in Tris-HCl 5OmM pH 7.4. This centrifugation step was repeated 3 times in total. After the last centrifugation step the pellet was resuspended in Tris-HCl 5OmM and homogenized with Dual Potter, 10 strokes to a homogenous solution, an aliquot was removed for protein determination. Membranes were aliquoted and frozen at -8O0C until use. The radioligand binding assay is performed at room temperature in 96-well microtiter plates (No-binding Surface Plates, Corning 3600) with a final assay volume of 200μl/well in incubation buffer (5OmM Tris buffer (pH 7.4 RT) containing 0.1 % BSA, 40 mg/L Bacitracin, complete EDTA-free protease inhibitor cocktail tablets 20 pills/L (Roche) and 3mM MnCl2). Competition binding curves were done by adding increasing amounts of the test compound. Test compounds were dissolved and serially diluted in DMSO, final
DMSO concentration 1.5 % in the assay. 50μl Non labelled ZD 6021 (a non selective NK- antagonist, lOμM final cone) was added for measurement of non-specific binding. For total binding, 50μl of 1.5% DMSO (final cone) in incubation buffer was used.[3H-Sar,Met(O2)- Substance P] (4nM final cone) was used in binding experiments on hNKir. [3H-SR48968] (3nM final cone.) for hNK2r and [3H-SRl 42801] (3nM final cone) for binding experiments on hNK3r. 50μl radioligand, 3μl test compound diluted in DMSO and 47 μl incubation buffer were mixed with 5-10μg cell membranes in lOOμl incubation buffer and incubated for 30 min at room temperature on a microplate shaker. The membranes were then collected by rapid filtration on Filtermat B(Wallac), presoaked in 0.1% BSA and 0.3% Polyethyleneimine (Sigma P-3143), using a Micro 96 Harvester (Skatron Instruments, Norway). Filters were washed by the harvester with ice-cold wash buffer (5OmM Tris-HCl, pH 7.4 at 4°C, containing 3mM MnCl2) and dried at 5O0C for 30- 60 min. Meltilex scintillator sheets were melted on to filters using a Microsealer (Wallac, Finland) and the filters were counted in a β-Liquid Scintillation Counter (1450 Microbeta, Wallac, Finland). The Kj value for the unlabeled ligand was calculated using the Cheng-Prusoff equation (Biochem. Pharmacol. 22:3099-3108, 1973): where L is the concentration of the radioactive ligand used and Kd is the affinity of the radioactive ligand for the receptor, determined by saturation binding. Data was fitted to a four-parameter equation using Excel Fit. K1 = IC50/ (1+(LZECd) )
Results
In general, the compounds of the invention, which were tested, demonstrated statistically significant antagonistic activity at the NKi receptor within the range of 8-9 for the pKg . For the NK2 receptor the range for the pKβ was 7-8. In general, the antagonistic activity at the NK3 receptor was 8-9 for the pKg.
In general, the compounds of the invention, which were tested, demonstrated statistically significant CYP3 A4 inhibition at a low level. The IC50 values tested according to Bapiro et al; Drug Metab. Dispos. 29, 30-35 (2001) were generally greater than 30 μM.
Activity against hERG
The activity of compounds according to formula I against the hERG-encoded potassium channel can be determined according to Kiss L, et al. Assay Drug Dev Technol. 1 (2003), 127-35: "High throughput ion-channel pharmacology: planar-array-based voltage clamp".
In general, the compounds of the invention, which were tested, demonstrated statistically significant hERG activity at a low level. The IC50 values tested as described above were generally greater than 5 μM. Metabolic stability
The metabolic stability of compounds according to formula I can be determined as described below:
The rate of biotransformation can be measured as either metabolite(s) formation or the rate of disappearance of the parent compound. The experimental design involves incubation of low concentrations of substrate (usually 1.0 μM) with liver microsomes (usually 0.5 mg/ml) and taking out aliquotes at varying time points (usually 0, 5, 10, 15, 20, 30, 40 min.). The test compound is usually dissolved in DMSO. The DMSO concentration in the
10 incubation mixture is usually 0.1% or less since more solvent can drastically reduce the activities of some CYP450s. Incubations are done in 100 mM potassium phosphate buffer, pH 7.4 and at 37 °C. Acetonitrile or methanol is used to stop the reaction. The parent compound is analysed by HPLC-MS. From the calculated half-life, t\a, the intrinsic clearance, Clint, is estimated by taking microsomal protein concentration and liver weight
I5 into account.
In general, the compounds of the invention had in vitro metabolic stability at a high level. Intrinsic clearance values tested as above were generally lower than 120 μl/min/mg protein.
20
The following tables illustrates the properties of the compounds of the present invention:
Methyl 4-{l-[(3S)-4-{[3-bromo-5-(trifluoromethyl)benzoyIJ(methyl)amino}-3-(4- uoro hen l but l azetidin-3- l i erazine-l-carbox late Ex 1
Figure imgf000023_0001
5 Methyl 4-{l-[(3S)-4-{[3-chloro-5-(trifluoromethyl)benzoyl](methyl)amino}-3-(4- uoro hen l but l 'azetidin-3- l i erazine-l-carbox late Ex 2
Figure imgf000024_0001
Biological evalution
GerbilFoot Tap (NKl specific test model)
Male Mongolian gerbils (60-8Og) are purchased from Charles River, Germany. On arrival, they are housed in groups often, with food and water ad libitum in temperature and humidity-controlled holding rooms. The animals are allowed at least 7 days to acclimatize to the housing conditions before experiments. Each animal is used only once and euthanized immediately after the experiment by heart punctuation or a lethal overdose of pentobarbital sodium.
Gerbils are anaesthetized with isoflurane. Potential CNS-permeable NKl receptor antagonists are administered intraperitoneally, intravenously or subcutaneously. The compounds are given at various time points (typically 30-120 minutes) prior to stimulation with agonist.
The gerbils are lightly anaesthetized using isofluorane and a small incision is made in the skin over bregma. 10 pmol of ASMSP, a selective NKl receptor agonist, is administered icv in a volume of 5 μl using a Hamilton syringe with a needle 4 mm long. The wound is clamped shut and the animal is placed in a small plastic cage and allowed to wake up. The cage is placed on a piece of plastic tubing filled with water and connected to a computer via a pressure transducer. The number of hind feet taps is recorded. Chrornodacryorrhea model (NKl specific test model)
The actions of antagonists at peripheral NKl receptors can be assessed in gerbils in vivo using the so-called chrornodacryorrhea model (Bristow LJ, Young L. Chrornodacryorrhea and repetitive hind paw tapping: models of peripheral and central tachykinin NKl receptor 5 activation in gerbils. Eur J Pharmacol 1994; 253: 245-252). Briefly, systemic (intravenous) administration of NKl receptor agonists to anaesthetized gerbils results in profuse secretion of red/brown tears in the eyes due to porphyrin secretion from the Harderian gland. NKl receptor antagonists block NKl -agonist evoked chromodacryorrhea. o Fecal pellet output (NK2 specific test model)
The in vivo effect (NK2) of the compounds of formula I can be determined by measuring NK2 receptor agonist-induced fecal pellet output using gerbil as described in e.g. The Journal of Pharmacology and Experimental Therapeutics (2001), pp. 559-564. 5
Colorectal distension model
Colorectal distension (CPJD) in gerbils is performed as previously described in rats and mice (Tammpere A, Brusberg M, Axenborg J, Hirsch I, Larsson H, Lindstrδm E. Evaluation of pseudo-affective responses to noxious colorectal distension in rats byo manometric recordings. Pain 2005; 116: 220-226; Arvidsson S, Larsson M, Larsson H, Lindstrδm E, Martinez V. Assessment of visceral pain-related pseudo-affective responses to colorectal distension in mice by intracolonic manometric recordings. J Pain 2006; 7: 108-118) with slight modifications. Briefly, gerbils are habituated to Bollmann cages 30- 60 min per day for three consecutive days prior to experiments to reduce motion artefactss due to restraint stress. A 2 cm polyethylene balloon (made in-house) with connecting catheter is inserted in the distal colon, 2 cm from the base of the balloon to the anus, during light isoflurane anaesthesia (Forene®, Abbott Scandinavia AB, Solna, Sweden). The catheter is fixed to the tail with tape. The balloons are connected to pressure transducers (P-602, CFM-k33, 100 mmHg, Bronkhorst HI-TEC, Veenendal, The Netherlands). Gerbils are allowed to recover from sedation in the Bollmann cages for at least 15 min before- the start of experiments.
A customized barostat (AstraZeneca, Mδlndal, Sweden) is used to manage air inflation and balloon pressure control. A customized computer software (PharmLab on-line 4.0) running on a standard computer is used to control the barostat and to perform data collection. The distension paradigm used consists of 12 repeated phasic distensions at 80 mmHg, with a pulse duration of 30 sec at 5 min intervals. Compounds or their respective vehicle are administered as intraperitoneal (i.p.) injections before the CRD paradigm. Each gerbil receives both vehicle and compound on different occasions with at least two days between experiments. Hence, each gerbil serves as its own vehicle control.
The analog input channels are sampled with individual sampling rates, and digital filtering is performed on the signals. The balloon pressure signals are sampled at 50 samples/s. A highpass filter at 1 Hz is used to separate the contraction-induced pressure changes from the slow varying pressure generated by the barostat. A resistance in the airflow between the pressure generator and the pressure transducer further enhances the pressure variations induced by abdominal contractions of the animal. A customized computer software (PharmLab off-line 4.0) is used to quantify the magnitude of highpass-filtered balloon pressure signals. The average rectified value (ARV) of the highpass-filtered balloon pressure signals is calculated for 30 s before the pulse (i.e baseline reponse) and for the duration of the pulse. When calculating the magnitude of the highpass-filtered balloon pressure signals, the first and last seconds of each pulse are excluded since these reflect artifact signals produced by the barostat during inflation and deflation and do not originate from the animal.

Claims

Claims
1. A compound of formula (I)
Figure imgf000027_0001
Q wherein
Ar is selected from
Figure imgf000027_0002
as well as pharmaceutically and pharmacologically acceptable salts thereof, and enantiomers of the compound of formula I and salts thereof.
2. A compound according to claim 1, wherein Ar is
Figure imgf000027_0003
3. A compound according to claim 1, wherein Ar is
Figure imgf000028_0001
5 '
4. A compound according to any one of claims 1-3 wherein the compound is the (S)- enantiomer.
5. A compound according to claim 1 selected from methyl 4-{l-[(35)-4-{[3-bromo-5- (trifluoromethyl)benzoyl](methyl)amino}-3-(4-fluorophenyl)butyl]azetidin-3-o yl}piperazine-l-carboxylate and Methyl 4-{l-[(35)-4-{[3-chloro-5-
(trifluoromethyl)benzoyl](methyl)amino}-3-(4-fluorophenyl)butyl]azetidin-3- yl}piperazine- 1 -carboxylate.
6. A compound according to any one of claims 1-5 for use in therapy. s
7. Use of a compound according to any one of claims 1-5 for the manufacture of a medicament for the treatment of a functional gastrointestinal disorder.
8. Use of a compound according to any one of claims 1-5 for the manufacture of ao medicament for the treatment of IBS.
9. Use of a compound according to any one of claims 1-5 for the manufacture of a medicament for the treatment of functional dyspepsia. 5
10. Use of a compound according to any one of claims 1-5 for the manufacture of a medicament for the treatment of IBD.
11. A pharmaceutical formulation comprising a compound according to any one of claims 1-5 as active ingredient and a pharmaceutically acceptable carrier or diluent.
12. A compound selected from Methyl 4-[l-(diphenylmethyl)azetidin-3-yl]piperazine-l-carboxylate; or
Methyl 4-azetidin-3-ylpiperazine-l -carboxylate.
PCT/SE2007/001115 2006-12-19 2007-12-18 Azetidinpiperazine derivatives that are neurokinin (nk) receptor antagonists and their use. WO2008076042A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US87064406P 2006-12-19 2006-12-19
US60/870,644 2006-12-19

Publications (1)

Publication Number Publication Date
WO2008076042A1 true WO2008076042A1 (en) 2008-06-26

Family

ID=39536552

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SE2007/001115 WO2008076042A1 (en) 2006-12-19 2007-12-18 Azetidinpiperazine derivatives that are neurokinin (nk) receptor antagonists and their use.

Country Status (1)

Country Link
WO (1) WO2008076042A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996005193A1 (en) * 1994-08-09 1996-02-22 Pfizer Limited (azetidin-1-ylalkyl)lactams as tachykinin antagonists
WO2004110344A2 (en) * 2003-06-13 2004-12-23 Astrazeneca Ab New azetidine compounds
WO2007037743A1 (en) * 2005-09-29 2007-04-05 Astrazeneca Ab Novel azetidine compounds useful in the treatment of functional gastrointestinal disorders, ibs and functional dyspepsia

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996005193A1 (en) * 1994-08-09 1996-02-22 Pfizer Limited (azetidin-1-ylalkyl)lactams as tachykinin antagonists
WO2004110344A2 (en) * 2003-06-13 2004-12-23 Astrazeneca Ab New azetidine compounds
WO2007037743A1 (en) * 2005-09-29 2007-04-05 Astrazeneca Ab Novel azetidine compounds useful in the treatment of functional gastrointestinal disorders, ibs and functional dyspepsia

Similar Documents

Publication Publication Date Title
US8288370B2 (en) Substituted azetidine compounds of formula (I) useful in the treatment of functional gastrointestinal disorders, IBS, and functional dyspepsia
US20100069346A1 (en) New Azetidine Derivatives as Neurokinin Receptor Antagonists for the Treatment of Gastrointestinal Diseases
WO2006137791A1 (en) New azetidine derivatives as neurokinin receptor antagonists for the treatment of gastrointestinal diseases
US8106208B2 (en) Benzamide compounds that act as NK receptor antagonists
EP1636221B1 (en) 3-Heterocyclyl-Azetidine compounds useful as NK1/NK2 receptors antagonists
WO2007037742A1 (en) Novel azetidine compounds useful in the treatment of functional gastrointestinal disorders, ibs and functional dyspepsia
US20080146538A1 (en) Compounds 614
WO2007136326A2 (en) ) t e text as een esta s e y t s ut o ty to rea as o ows: 1- [ (4- [benzoyl (methyl) amino] -3- (phenyl) butyl] azetidine derivatives for the treatment of gastrointestinal disorders 3
US20070270400A1 (en) New compounds 303
WO2008076042A1 (en) Azetidinpiperazine derivatives that are neurokinin (nk) receptor antagonists and their use.
MX2008004263A (en) Novel azetidine compounds useful in the treatment of functional gastrointestinal disorders, ibs and functional dyspepsia

Legal Events

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

Ref document number: 07852117

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07852117

Country of ref document: EP

Kind code of ref document: A1