US20090170869A1 - Piperazine derivative having affinity for the histamine h3 receptor - Google Patents

Piperazine derivative having affinity for the histamine h3 receptor Download PDF

Info

Publication number
US20090170869A1
US20090170869A1 US12/203,946 US20394608A US2009170869A1 US 20090170869 A1 US20090170869 A1 US 20090170869A1 US 20394608 A US20394608 A US 20394608A US 2009170869 A1 US2009170869 A1 US 2009170869A1
Authority
US
United States
Prior art keywords
tetrahydro
yloxy
pyran
carbonyl
phenyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/203,946
Other languages
English (en)
Inventor
Desmond John Best
Sing Yeung Mak
Barry Sidney Orlek
Geracimos Rassias
Pamela Joan Theobald
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Glaxo Group Ltd
Original Assignee
Glaxo Group Ltd
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=39885024&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20090170869(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from GB0717336A external-priority patent/GB0717336D0/en
Priority claimed from GB0814987A external-priority patent/GB0814987D0/en
Application filed by Glaxo Group Ltd filed Critical Glaxo Group Ltd
Publication of US20090170869A1 publication Critical patent/US20090170869A1/en
Assigned to GLAXO GROUP LIMITED reassignment GLAXO GROUP LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THEOBALD, PAMELA JOAN, BEST, DESMOND JOHN, MAK, SING YEUNG, ORLEK, BARRY SIDNEY, RASSIAS, GERACIMOS
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D309/08Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D309/10Oxygen atoms
    • C07D309/12Oxygen atoms only hydrogen atoms and one oxygen atom directly attached to ring carbon atoms, e.g. tetrahydropyranyl ethers
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention relates to a novel piperazine derivative having pharmacological activity, to processes for its preparation, to compositions containing it, and to its use in the treatment of neurological or psychiatric disorders such as cognitive impairment e.g. in Alzheimer's disease.
  • the histamine H3 receptor is predominantly expressed in the mammalian central nervous system (CNS), with minimal expression in peripheral tissues except on some sympathetic nerves (Leurs et al., (1998), Trends Pharmacol. Sci. 19, 177-183). Activation of H3 receptors by selective agonists or histamine results in the inhibition of neurotransmitter release from a variety of different nerve populations, including histaminergic and cholinergic neurons (Schlicker et al., (1994), Fundam. Clin. Pharmacol. 8, 128-137).
  • H3 antagonists can facilitate neurotransmitter release in brain areas such as the cerebral cortex and hippocampus, relevant to cognition (Onodera et al., (1998), In: The Histamine H3 receptor, ed. Leurs and Timmerman, pp 255-267, Elsevier Science B.V.).
  • H3 antagonists e.g. thioperamide, clobenpropit, ciproxifan and GT-2331
  • rodent models including the five choice task, object recognition, elevated plus maze, acquisition of novel task and passive avoidance (Giovanni et al., (1999), Behav. Brain Res.
  • the histamine H3 receptor antagonist GSK189254 inhibited [3H]R- ⁇ -methylhistamine ex vivo binding in the rat cortex following oral administration to the rat, and at certain oral doses improved performance of rats in the following cognition paradigms: passive avoidance, water maze, object recognition, and attentional set shift (A. D. Medhurst et al., J. Pharmacol. Exp. Therap., 2007, 321(3), 1032-1045).
  • H3 antagonists and/or inverse agonists could be useful for the treatment of cognitive impairments in neurological diseases such as Alzheimer's disease or a related neurodegenerative disorder.
  • WO 2005/040144 A1 (Glaxo Group Limited) discloses a series of 1-benzoyl-substituted diazepanyl derivatives having affinity for and being antagonists and/or inverse agonists of the histamine H3 receptor.
  • Example 10 of WO 2005/040144 A1 discloses 1-(isopropyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ hexahydro-1H-1,4-diazepine hydrochloride:
  • WO 2004/037801 A1 Janssen Pharmaceutica, N.V. discloses a series of piperazinyl and diazepanyl benzamides and benzothiamides with the ability to modulate the activity of the histamine receptor, specifically the H3 receptor.
  • WO 2004/101546 A1 discloses a number of (piperidine-4-carbonyl)-piperazine derivatives and (piperidine-4-carbonyl)-[1,4]-diazepane derivatives having affinity for and being antagonists and/or inverse agonists of the histamine H3 receptor.
  • WO 03/004480 A2 (Novo Nordisk A/S and Boehringer Ingelheim International GmbH) discloses a series of substituted piperazines and diazapanes having binding affinity to the histamine H3 receptor.
  • the present invention provides a compound or salt thereof, which has affinity for, and which is an antagonist and/or inverse agonist of, the histamine H3 receptor.
  • the present invention provides, in a first aspect, 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine
  • FIG. 1 is an X-ray powder diffraction (XRPD) spectrum of crystalline Form 1 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, expressed in terms of two-theta angles (in degrees), and obtained with a diffractometer using copper K ⁇ (copper K-alpha) X-radiation, with a step size of 0.01670 two-theta, a time per step of 31.75 sec, and using a sample mounted on a silicon wafer plate.
  • XRPD X-ray powder diffraction
  • FIG. 2 is an X-ray powder diffraction (XRPD) spectrum of crystalline Form 2 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, expressed in terms of two-theta angles (in degrees), and obtained with a diffractometer using copper K ⁇ (copper K-alpha) X-radiation, with a step size of 0.01670 two-theta, a time per step of 31.75 sec, and using a sample mounted on a silicon wafer plate.
  • XRPD X-ray powder diffraction
  • FIG. 3 is an overlay of a portion of the XRPD spectra for crystalline Form 1 (top) and crystalline Form 2 (bottom) of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride.
  • FIG. 4 is a Fourier-Transform Infrared (FT-IR) spectrum for crystalline Form 1 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, showing the spectral region from 4000 to 675 cm ⁇ 1 .
  • FT-IR Fourier-Transform Infrared
  • FIG. 5 is a Fourier-Transform Infrared (FT-IR) spectrum for crystalline Form 1 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, showing the spectral region from 2000 to 675 cm ⁇ 1 .
  • FT-IR Fourier-Transform Infrared
  • FIG. 6 is a Fourier-Transform Infrared (FT-IR) spectrum for crystalline Form 2 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, showing the spectral region from 4000 to 675 cm ⁇ 1 .
  • FT-IR Fourier-Transform Infrared
  • FIG. 7 is a Fourier-Transform Infrared (FT-IR) spectrum for crystalline Form 2 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, showing the spectral region from 2000 to 675 cm ⁇ 1 .
  • FT-IR Fourier-Transform Infrared
  • FIG. 8 is an overlay of the FT-IR spectra for crystalline Form 1 (top) and crystalline Form 2 (bottom) of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, showing the spectral region from 2000 to 675 cm ⁇ 1 .
  • FIG. 9 is an overlay of the 13 C solid-state nuclear magnetic resonance (solid-state NMR) spectra, in ppm, of crystalline Form 1 (top) and crystalline Form 2 (bottom) of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride.
  • FIG. 10 is a scheme showing how the receptor occupancy of a test compound (“drug candidate”) can be measured in vivo by the reduction in radioligand specific binding to receptors.
  • B A is the concentration of available receptor sites. Notice how B A changes between baseline, and 10 min, 2.5 hours, and 6 hours after administration of the test compound, as a consequence of the presence of different concentrations of the drug candidate in tissue.
  • FIG. 11 is a scheme showing the pig-PET protocol to measure the H3 receptor occupancy of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride (“salt A”, within the present invention), and 1-(isopropyl)-4-[4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonylhexahydro-1H-1,4-diazepine hydrochloride (“salt B”, a comparator compound).
  • FIG. 12 is a graph showing the average (mean) plasma concentration over time of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride (“salt A”, within the present invention, filled circles) and 1-(isopropyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ hexahydro-1H-1,4-diazepine hydrochloride (“salt B”, a comparator compound, filled diamonds), following 50 micrograms/kg intravenous administration to pigs.
  • salt A 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride
  • salt B a comparator compound, filled diamonds
  • FIG. 12 is a graph showing the average (mean) measured H3 receptor occupancy time course at three time points during an in vivo pig-PET study, and the k on -k off limited model fitted to it, for “salt A” within the present invention (measurements as filled circles, and model fit as solid line), and for “salt B” a comparator compound (measurements as filled diamonds, and model fit as dashed line), following 50 micrograms/kg intravenous administration of salt A or salt B to pigs.
  • FIG. 13 parts A and B, are graphs showing the average (mean) plasma concentration over time and average (mean) H3 receptor occupancy time course respectively for “salt A” (only), as shown in part of FIG. 12 graphs A and B.
  • FIG. 13 parts C and D, are graphs showing the average (mean) plasma concentration over time and average (mean) H3 receptor occupancy time course respectively for “salt B” (only), as shown in part of FIG. 12 graphs A and B.
  • the present invention provides a compound or salt thereof, which has affinity for, and which is an antagonist and/or inverse agonist of, the histamine H3 receptor.
  • the present invention provides, in a first aspect, 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine
  • 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine has exhibited in rats and pigs certain time courses (decays over time) of brain histamine H3 receptor occupancy (see the Rat ex vivo binding studies and the Pig-PET studies hereinafter), which suggest that 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof might have certain suitable properties for human pharmaceutical use, in particular in the treatment of cognitive impairment in humans such as cognitive impairment in Alzheimer's disease.
  • reference to 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine (the “free base”) or a salt thereof encompasses solvates and hydrates of the free base or the salt thereof.
  • the invention provides 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine (the “free base”).
  • a salt of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine is preferably a pharmaceutically acceptable salt thereof, in particular a pharmaceutically acceptable acid addition salt thereof.
  • compositions of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine include hydrobromide (e.g. monohydrobromide), hydrochloride (e.g.
  • salts can generally be formed by mixing with the appropriate acid, optionally in a suitable solvent such as an organic solvent, to give the salt, which can be isolated, for example by crystallisation and filtration, usually followed by drying.
  • the invention includes within its scope all possible stoichiometric and non-stoichiometric forms of the salts of the compound of the invention including hydrates and solvates.
  • the compound or salt is in the form of a hydrochloride salt, 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, typically the monohydrochloride salt.
  • 1-(1-Methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride e.g. monohydrochloride
  • the invention therefore also provides crystalline Form 1 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, e.g. monohydrochloride, (hereinafter “crystalline Form 1”).
  • Crystalline Form 1 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride e.g. monohydrochloride
  • XRPD X-ray powder diffraction
  • crystalline Form 1 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride e.g. monohydrochloride
  • XRPD X-ray powder diffraction
  • crystalline Form 1 characterised by the XRPD spectrum peaks defined herein and/or characterised by an XRPD spectrum substantially the same as that shown in FIG. 1 , can be additionally characterised as having been obtained with a diffractometer using a step size of 0.0167° two-theta or less, and/or a time per step of 31.75 sec or more, and/or using a sample mounted on a silicon wafer plate.
  • crystalline Form 1 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride can be characterised by a solid-form Fourier-Transform Infrared (FT-IR) spectrum substantially the same as that shown in FIG. 5 .
  • FIG. 5 shows the FT-IR spectrum of crystalline Form 1 in the spectral region from 2000 to 675 cm ⁇ 1 .
  • the FT-IR spectrum can e.g. be measured using a Nicolet Avatar 360 FT-IR spectrometer, and/or can e.g. be as measured at 4 cm ⁇ 1 or 2 cm ⁇ 1 resolution.
  • a variation can be allowed for each peak of about +2 cm ⁇ 1 .
  • crystalline Form 1 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride e.g.
  • monohydrochloride can be characterised by a 13 C solid-state nuclear magnetic resonance (solid-state NMR) spectrum having the following chemical shifts for the resonances: 18.5 ⁇ 0.3, 30.4 ⁇ 0.3, 31.8 ⁇ 0.3, 37.6 ⁇ 0.3, 45.8 ⁇ 0.3, 49.4 ⁇ 0.3, 52.3 ⁇ 0.3, 59.2 ⁇ 0.3, 63.6 ⁇ 0.3, 68.4 ⁇ 0.3, 110.3 ⁇ 0.3, 118.8 ⁇ 0.3, 128.4 ⁇ 0.3, 131.2 ⁇ 0.3, 133.9 ⁇ 0.3, 159.1 ⁇ 0.3, and 167.6 ⁇ 0.3 ppm.
  • This solid-state NMR spectrum can for example be obtained at a frequency of 90.55 MHz for 13 C observation, e.g.
  • Bruker HFX MAS magic-angle spinning
  • Data can e.g. be acquired using a cross polarisation sequence with side-band suppression.
  • a relaxation delay of 10 seconds can be used during scanning.
  • the hydrochloride salt of the invention is substantially (e.g. 60% or more or 70% or more or 80% or more by weight or molarity) in the form of crystalline Form 1 in terms of crystal form purity.
  • the invention also provides crystalline Form 2 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, e.g. monohydrochloride, (hereinafter “crystalline Form 2”).
  • crystalline Form 2 appears to be more thermodynamically stable than crystalline Form 1, which may give certain advantages in relation to storage, formulation and/or use.
  • Crystalline Form 2 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride e.g. monohydrochloride
  • XRPD X-ray powder diffraction
  • crystalline Form 2 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride e.g. monohydrochloride
  • XRPD X-ray powder diffraction
  • crystalline Form 2 characterised by the XRPD spectrum peaks defined herein and/or characterised by an XRPD spectrum substantially the same as that shown in FIG. 2 , can be additionally characterised as having been obtained with a diffractometer using a step size of 0.0167° two-theta or less, and/or a time per step of 31.75 sec or more, and/or using a sample mounted on a silicon wafer plate.
  • crystalline Form 2 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride can be characterised by a solid-form Fourier-Transform Infrared (FT-IR) spectrum substantially the same as that shown in FIG. 7 .
  • FIG. 7 shows the FT-IR spectrum of crystalline Form 2 in the spectral region from 2000 to 675 cm ⁇ 1 .
  • the FT-IR spectrum can e.g. be measured using a Nicolet Avatar 360 FT-IR spectrometer, and/or can e.g. be as measured at 4 cm ⁇ 1 or 2 cm ⁇ 1 resolution.
  • a variation can be allowed for each peak of ⁇ 2 cm ⁇ 1 such as ⁇ 1 cm ⁇ 1 .
  • crystalline Form 2 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride e.g.
  • monohydrochloride can be characterised by a 13 C solid-state nuclear magnetic resonance (solid-state NMR) spectrum having the following chemical shifts for the resonances: 18.8 ⁇ 0.3, 19.5 ⁇ 0.3, 32.4 ⁇ 0.3, 37.5 ⁇ 0.3, 45.7 ⁇ 0.3, 49.3 ⁇ 0.3, 52.7 ⁇ 0.3, 59.1 ⁇ 0.3, 66.3 ⁇ 0.3, 71.1 ⁇ 0.3, 109.4 ⁇ 0.3, 119.6 ⁇ 0.3, 128.4 ⁇ 0.3, 131.3 ⁇ 0.3, 134.3 ⁇ 0.3, 158.7 ⁇ 0.3, and 167.8 ⁇ 0.3 ppm.
  • This solid-state NMR spectrum can for example be obtained at a frequency of 90.55 MHz for 13 C observation, e.g.
  • Bruker HFX MAS magic-angle spinning
  • Data can e.g. be acquired using a cross polarisation sequence with side-band suppression.
  • a relaxation delay of 10 seconds can be used during scanning.
  • the hydrochloride salt of the invention can suitably be substantially (e.g. 70% or more or 80% or more or 90% or more or 95% or more by weight or molarity) in the form of crystalline Form 2 in terms of crystal form purity.
  • the present invention also provides a process for the preparation of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a salt (e.g. pharmaceutically acceptable salt) thereof, which process comprises:
  • Process (a) typically comprises the use of amide formation conditions in the presence of a suitable base such as triethylamine or a solid supported base (e.g. diethylaminomethylpolystyrene), in an appropriate solvent e.g. a non-aqueous organic solvent such as dichloromethane, at an appropriate temperature, for example from about ⁇ 10° C. to about 40° C., such as room temperature.
  • a catalytic amount of N,N-dimethylformamide (DMF) is added to catalyse the reaction.
  • room temperature is usually 12-35° C., for example 18-30° C. or 18-25° C., such as about 22° C.
  • Process (b) typically comprises activation of 4-(tetrahydro-2H-pyran-4-yloxy)benzoic acid with a coupling reagent, e.g. in a suitable solvent e.g. a polar aprotic organic solvent, such as N,N-dimethylformamide, dimethylsulfoxide, acetonitrile or propionitrile, followed by reaction with 1-isopropyl piperazine.
  • a suitable solvent e.g. a polar aprotic organic solvent, such as N,N-dimethylformamide, dimethylsulfoxide, acetonitrile or propionitrile
  • the coupling reagent is an organic di-substituted carbodiimide, such as 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC) or dicyclohexylcarbodiimide (DCC), in which case: the reaction can optionally be carried out in the presence of 1-hydroxybenzotriazole (HOBT), and/or the reaction solvent can for example be N,N-dimethylformamide, and/or the reaction temperature can e.g. be from about 0° C. to about 40° C., such as room temperature.
  • EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • DCC dicyclohexylcarbodiimide
  • the reaction can optionally be carried out in the presence of 1-hydroxybenzotriazole (HOBT), and/or the reaction solvent can for example be N,N-dimethylformamide, and/or the reaction temperature can
  • the coupling reagent is carbonyl diimidazole, pivaloyl chloride (trimethylacetyl chloride) or 2-propane phosphonic acid anhydride.
  • the coupling reagent is carbonyl diimidazole (CDI).
  • reaction solvent comprising (or, in one particular embodiment, consisting essentially of) acetonitrile and/or propionitrile, more preferably acetonitrile.
  • reaction conditions can in particular be as follows, independently and/or in any combination:
  • a salt e.g. hydrochloride
  • a coupling reagent e.g. carbonyl diimidazole
  • reaction of the activated acid with the 1-isopropyl piperazine in a reaction solvent such as acetonitrile or propionitrile, the following process can be carried out:
  • the solvent-containing mixture comprising the salt (e.g. HCl salt) product is at, or is heated to, a temperature of about 50° C. to the boiling point or reflux temperature of the solvent (e.g. about 50-75° C., e.g. about 60-70° C., e.g. about 60-65° C.), and
  • the salt e.g. hydrochloride salt
  • the salt of the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine is allowed or caused to crystallise or recrystallise from the hot mixture (e.g. by cooling the hot mixture), and
  • hydrochloride e.g. monohydrochloride
  • 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine without being bound be theory, it appears, from preliminary experiments on the above-mentioned types of HCl-salt-formations and crystallisations, that crystalline Form 1 often tends to be the kinetic product of the salt-forming process, and that crystalline Form 2 is the thermodynamic product (i.e. thermodynamically more stable product).
  • Crystalline Form 1 (or a predominance of crystalline Form 1) is often formed initially depending on the conditions, but, depending on the conditions (such as the type of solvent and the solubility of crystalline Form 1 in it, and/or the temperature and/or temperature time course, and/or the contact time of the crystalline Form 1 with the solvent), the crystalline Form 1 can then often convert to crystalline Form 2 to a greater or lesser extent when in contact with a suitable solvent i.e. a solvent suitable for converting crystalline Form 1 to crystalline Form 2.
  • a suitable solvent i.e. a solvent suitable for converting crystalline Form 1 to crystalline Form 2.
  • the invention in one aspect, therefore provides a process for preparing crystalline Form 2 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride (e.g. monohydrochloride), comprising: converting crystalline Form 1 to crystalline Form 2 by contacting crystalline Form 1 with a suitable conversion solvent, for example by slurrying crystalline Form 1 in the suitable conversion solvent, for a sufficient time and/or at a sufficiently-high temperature to effect conversion of crystalline Form 1 to crystalline Form 2.
  • the suitable conversion solvent typically comprises (e.g.
  • the crystalline product of the process is suitably substantially (e.g. 70% or more or 80% or more or 90% or more or 95% or more by weight or molarity) in the form of crystalline Form 2 in terms of crystal form purity.
  • 4-(Tetrahydro-2H-pyran-4-yloxy)benzoyl chloride (V) or 4-(tetrahydro-2H-pyran-4-yloxy)benzoic acid (IV) may be prepared in accordance with the following scheme wherein P represents a suitable protecting group, such as C 1-6 straight-chain alkyl (e.g. methyl, ethyl, n-propyl or n-butyl) or isopropyl or isobutyl, or benzyl; such as methyl or ethyl; in particular methyl.
  • P represents a suitable protecting group, such as C 1-6 straight-chain alkyl (e.g. methyl, ethyl, n-propyl or n-butyl) or isopropyl or isobutyl, or benzyl; such as methyl or ethyl; in particular methyl.
  • Step (i) typically comprises the use of a phosphine such as triphenylphosphine in a suitable solvent such as tetrahydrofuran, toluene and/or xylene (wherein “xylene” can be o-xylene, m-xylene, p-xylene, or a mixture of xylenes), followed by the addition (e.g. slow and/or dropwise addition) of an azodicarboxylate such as diethyl azodicarboxylate or diisopropyl azodicarboxylate, at a suitable temperature, for example, from room temperature to about 80° C., e.g. room temperature.
  • a suitable solvent such as tetrahydrofuran, toluene and/or xylene
  • xylene can be o-xylene, m-xylene, p-xylene, or a mixture of xylenes
  • Reaction times can be e.g. from 0.5 to 72 hours.
  • room temperature can be used, and the reaction time is for example from 3 to 72 hours.
  • the reaction solvent comprises or consists essentially of toluene and/or xylene, in particular toluene
  • a reaction temperature of about 40 to about 80° C. e.g. about 40 to about 70° C., e.g. about 55° C.
  • a reaction time (including any azodicarboxylate addition time) of about 0.5 to 6 hours, e.g. 0.5 to 3 hours, e.g. 1-2 hours, can be used.
  • the reaction solvent comprises or consists essentially of toluene and/or xylene, preferably toluene
  • reaction step (i) uses triphenylphosphine and diisopropyl azodicarboxylate; in which case suitably the heated (e.g. ca. 40-70° C.) reaction mixture can be cooled (e.g. to ⁇ 10 to 25° C., e.g. to ca. 0-5° C., provided that it is not cooled to the melting point of the solvent or below), e.g. for 0.5 to 2 hours, and then the solid byproduct formed is removed e.g. by filtration.
  • toluene helps to crystallise out the byproduct adduct of triphenylphosphine oxide and diisopropyl hydrazinedicarboxylate from the solution (especially when the reaction mixture is seeded with this adduct e.g. after cooling), which helps to reduce the levels of triphenylphosphine oxide in the crude product (III).
  • reaction product compound of formula (III) is not isolated.
  • the toluene and/or xylene solution of the compound of formula (III) is used directly in the subsequent reaction (deprotection e.g. hydrolysis) step (ii), in particular when C 1-6 straight-chain alkyl (e.g. methyl, ethyl, n-propyl or n-butyl) or isopropyl or isobutyl and the subsequent step (ii) comprises alkaline (e.g. NaOH or KOH) hydrolysis of the ester.
  • alkaline e.g. NaOH or KOH
  • P represents a protecting group such as C 1-6 straight-chain alkyl (e.g. methyl, ethyl, n-propyl or n-butyl) or isopropyl or isobutyl, or benzyl (in particular C 1-6 straight-chain alkyl or isopropyl, e.g. methyl or ethyl), wherein the process comprises:
  • P represents the protecting group as defined for the compound of formula (III), with 4-hydroxytetrahydropyran of formula (II) or a derivative thereof in which its OH group is activated;
  • Step (ii) is a deprotection reaction.
  • P represents C 1-6 straight-chain alkyl (e.g. methyl, ethyl, n-propyl or n-butyl) or isopropyl or isobutyl (in particular methyl or ethyl)
  • a suitable alkali e.g. aqueous
  • sodium hydroxide or potassium hydroxide e.g. aqueous sodium hydroxide or potassium hydroxide solution
  • reaction solvent is toluene and/or xylene
  • the reaction comprises treatment with a suitable aqueous alkali such as aqueous sodium hydroxide or potassium hydroxide solution
  • the reaction comprises efficient (e.g. vigorous) stirring or mixing.
  • a toluene and/or xylene solution containing the compound of formula (III), produced in step (i), is used directly in the subsequent hydrolysis step (ii), i.e. without isolation of the compound of formula (III), in particular when the subsequent step (ii) comprises alkaline (e.g. NaOH or KOH) hydrolysis of the ester.
  • alkaline e.g. NaOH or KOH
  • the reaction conditions for steps (i) and/or (ii) can in particular be as described herein, e.g. reaction step (i) can use triphenylphosphine and diisopropyl azodicarboxylate.
  • the deprotection reaction (ii) can comprise hydrogenation.
  • reaction steps (i) and (ii) are both carried out in a reaction solvent comprising or consisting essentially of toluene and/or xylene (in particular toluene).
  • Xylene can be o-xylene, m-xylene, p-xylene, or a mixture of xylenes.
  • the toluene and/or xylene solution of the compound of formula (III) produced in step (i) is used directly in the subsequent hydrolysis step (ii), i.e. without isolation of the compound of formula (III), in particular when the subsequent step (ii) comprises alkaline (e.g. NaOH or KOH) hydrolysis of the ester.
  • alkaline e.g. NaOH or KOH
  • the reaction conditions for steps (i) and/or (ii) can in particular be as described herein, e.g. reaction step (i) can use triphenylphosphine and diisopropyl azodicarboxylate.
  • Steps a) and/or b) can e.g. be as described herein.
  • Step (iii) typically comprises treatment with suitable chlorinating agent such as oxalyl chloride or thionyl chloride, e.g. in a suitable solvent (e.g. non-aqueous organic solvent) such as dichloromethane or ethyl acetate (suitably dichloromethane), or (for thionyl chloride) without solvent, at a suitable temperature, such as room temperature.
  • suitable chlorinating agent such as oxalyl chloride or thionyl chloride
  • suitable solvent e.g. non-aqueous organic solvent
  • suitable temperature such as room temperature.
  • 1-(1-Methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof has affinity for and is an antagonist and/or inverse agonist of the histamine H3 receptor, and for example has potentially useful therapeutic properties.
  • 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof has potential use in the treatment or prophylaxis (in particular treatment) of:
  • the invention also provides 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof, for use as a therapeutic substance in the treatment or prophylaxis (in particular treatment) of any of the above disorders; in particular cognitive impairment(s), e.g. cognitive impairment(s) in a disease such as Alzheimer's disease, dementia (e.g. Lewy body dementia or vascular dementia), mild cognitive impairment, or a related neurodegenerative disorder, or cognitive impairment(s) in Parkinson's disease, or cognitive impairment(s) in schizophrenia; or fatigue; or a sleep disorder.
  • cognitive impairment(s) e.g. cognitive impairment(s) in a disease such as Alzheimer's disease, dementia (e.g. Lewy body dementia or vascular dementia), mild cognitive impairment, or a related neurodegenerative disorder, or cognitive impairment(s) in Parkinson's disease, or cognitive impairment(s) in schizophrenia; or fatigue; or
  • the invention also provides 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof, for use as a therapeutic substance in the treatment or prophylaxis (in particular treatment) of any of the above disorders, in particular cognitive impairment(s), fatigue or a sleep disorder, in a mammal (e.g. rodent such as rat, or pig or human) such as a human.
  • a mammal e.g. rodent such as rat, or pig or human
  • the invention further provides a method of treatment or prophylaxis (in particular treatment) of any of the above disorders, e.g. a neurological disease, in a mammal such as a human, which comprises administering to the sufferer (the mammal in need thereof) a therapeutically effective amount of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof.
  • the invention further provides a method of treatment or prophylaxis (in particular treatment) of:
  • the invention provides the use of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for use in the treatment or prophylaxis (in particular treatment) of any of the above disorders, in particular a neurological disease and/or in particular cognitive impairment(s), fatigue or a sleep disorder.
  • the invention provides the use of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for use in the treatment or prophylaxis (in particular treatment) of any of the above disorders, in particular a neurological disease and/or in particular cognitive impairment(s), fatigue or a sleep disorder, in a mammal (e.g. rodent such as rat, or pig or human) such as a human.
  • a mammal e.g. rodent such as rat, or pig or human
  • the invention provides the use of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof in the manufacture of a medicament:
  • the compound of the invention or a pharmaceutically acceptable salt thereof is usually formulated in a pharmaceutical composition.
  • Such compositions can be prepared using various procedures.
  • the present invention further provides a pharmaceutical composition for use in the treatment or prophylaxis (e.g. treatment) of any the above disorders, e.g. a neurological disease and/or cognitive impairment(s), fatigue or a sleep disorder, which comprises 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition for use in the treatment or prophylaxis (e.g. treatment) of any the above disorders e.g. a neurological disease and/or cognitive impairment(s), fatigue or a sleep disorder, which comprises 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • the present invention further provides a pharmaceutical composition which comprises 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition of the invention which may be prepared by admixture, for example at ambient temperature and/or atmospheric pressure, is usually adapted for oral, parenteral or rectal administration and, as such, may be in the form of a tablet, a capsule, an oral liquid preparation, a powder, granules, a lozenge, a reconstitutable powder, an injectable or infusible solution or suspension, or a suppository.
  • An orally administrable pharmaceutical composition such as a tablet or capsule, is generally preferred.
  • a tablet or capsule for oral administration may be in unit dose form, and may contain one or more excipients, such as a binding agent (e.g. povidone, hydroxypropylmethylcellulose or starch), a filler (e.g. mannitol or lactose), microcrystalline cellulose, a lubricant e.g. tabletting lubricant (e.g. magnesium stearate, calcium stearate or stearic acid), a disintegrant e.g. tablet disintegrant, and/or a pharmaceutically acceptable wetting agent.
  • a tablet may be coated, e.g. film-coated, e.g. according to a tablet coating method.
  • a capsule can be a hard or soft capsule, containing the compound or salt of the invention and the one or more excipients e.g. in powder or pellet form.
  • An oral liquid preparation may be in the form of, for example, an aqueous or oily suspension, a solution, an emulsion, a syrup or elixir, or may be in the form of a dry product for reconstitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain additive(s) such as suspending agents, emulsifying agents, non-aqueous vehicles (which may include edible oils), and/or preservatives, and/or, if desired, flavourings and/or colorants.
  • fluid unit dosage forms are typically prepared utilising the compound of the invention or pharmaceutically acceptable salt thereof and a sterile vehicle.
  • the compound or salt e.g. depending on the vehicle and/or concentration used, can be either suspended or dissolved in the vehicle.
  • the compound or salt can be dissolved for injection and filter sterilised before filling into a suitable vial or ampoule and sealing.
  • Adjuvant(s) such as a local anaesthetic, preservative(s) and/or buffering agent(s) can be dissolved in the vehicle.
  • the composition can be frozen after filling into the vial and the water removed under vacuum.
  • Parenteral suspensions are typically prepared in substantially the same manner, except that the compound or salt is suspended in the vehicle instead of being dissolved, and sterilisation typically is not accomplished by filtration.
  • the compound or salt is sterilised, e.g. by exposure to ethylene oxide, before suspension in a sterile vehicle.
  • a surfactant or wetting agent is included in the composition to facilitate uniform distribution of the compound or salt.
  • the pharmaceutical composition may contain from 0.1% to 99% by weight of the composition of the active material (i.e. the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the pharmaceutically acceptable salt thereof), in particular from 1 to 60% by weight or from 10 to 60% by weight of the composition of the active material. For example, this may vary depending on the route of administration and/or the composition's intended use(s).
  • the composition of the active material i.e. the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the pharmaceutically acceptable salt thereof.
  • this may vary depending on the route of administration and/or the composition's intended use(s).
  • the total amount of the pharmaceutically acceptable carrier in the pharmaceutical composition can for example vary depending on the pharmaceutical composition and/or its intended use and/or the route of administration.
  • the total amount of the pharmaceutically acceptable carrier in the pharmaceutical composition e.g. or i.e. the total amount of the one or more excipients present therein, such as one or more of the excipient types mentioned herein
  • the total amount of the pharmaceutically acceptable carrier in the unit dose form pharmaceutical composition can be from 10 mg to 2000 mg, for example from 20 mg to 1500 mg such as from 100 mg to about 1000 mg.
  • the dose, e.g. oral dose, of the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the pharmaceutically acceptable salt thereof, e.g. used in the treatment or prophylaxis of the aforementioned disorders/diseases /conditions and/or comprised in a pharmaceutical composition can for example vary in the usual way with the seriousness of the disorders, the weight of the sufferer, and/or other similar factors.
  • a suitable unit dose e.g.
  • oral unit dose of 0.02 to 1000 mg or 0.05 to 1000 mg, for example 0.1 to 200 mg such as 1.0 to 200 mg, and/or for example 0.02 to 200 mg or 0.05 to 200 mg such as 0.05 to 45 mg or 0.1 to 45 mg, of the compound or the pharmaceutically acceptable salt of the invention (measured as the “free base” compound), may be used, for example in a pharmaceutical composition (e.g. in an oral pharmaceutical composition, and/or e.g. in a unit dose form) of the invention.
  • a unit dose is for administration once a day, e.g.
  • Such a unit dose may be for administration more than once a day, for example two or three times a day, e.g. orally and/or to a mammal such as a human.
  • Such therapy may extend for a number of weeks, months or years.
  • the invention provides a pharmaceutical dosage form (e.g. orally-administrable dosage form) comprising:
  • a stabiliser which reduces degradation of the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the salt thereof in the dosage form when compared to a dosage form lacking said stabiliser.
  • the pharmaceutical dosage form (e.g. orally-administrable dosage form) comprises a carrier tablet, which carrier tablet is at least partially (e.g. partially or wholly, e.g. only partially) covered by a film comprising:
  • a stabiliser that reduces degradation of the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the salt thereof in the dosage form, when compared to a dosage form lacking said stabiliser.
  • carrier tablet refers to a pharmaceutically acceptable tablet substantially free of the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the pharmaceutically acceptable salt thereof.
  • the carrier tablet is formed by direct compression technology.
  • the carrier tablet comprises:
  • the carrier tablet is a tablet comprising microcrystalline cellulose (e.g. Avicel PH-102 TM) (e.g. at 90% by weight of the carrier tablet), pregelatinized starch (e.g. Starch 1500 TM) (e.g. at 9% by weight of the carrier tablet), and magnesium stearate (e.g. at 1% by weight of the carrier tablet).
  • microcrystalline cellulose e.g. Avicel PH-102 TM
  • pregelatinized starch e.g. Starch 1500 TM
  • magnesium stearate e.g. at 1% by weight of the carrier tablet.
  • the above-mentioned pharmaceutical dosage form comprising the optional stabiliser can for example contain from 0.02 mg to 2 mg (e.g. 0.05 mg to 1 mg) of the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the pharmaceutically acceptable salt thereof (e.g. hydrochloride salt), when measured as the amount of free base present.
  • 0.02 mg to 2 mg e.g. 0.05 mg to 1 mg
  • the pharmaceutically acceptable salt thereof e.g. hydrochloride salt
  • the dosage form does not comprise a stabiliser.
  • the dosage form does comprise a stabiliser.
  • the stabiliser can typically comprise citric acid or a salt thereof, malic acid or a salt thereof, ascorbic acid or a salt thereof, sodium bicarbonate, optionally butylated hydroxyanisole and/or butylated hydroxytoluene.
  • the stabiliser comprises optionally butylated hydroxyanisole, such as butylated hydroxyanisole, or, more particularly, citric acid or a salt thereof, such as citric acid.
  • the molar ratio of the citric acid or the salt thereof (measured as citric acid) to the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the salt thereof (measured as the free base) can for example be from 550:1 to 1:2, such as from 500:1 to 2:3.
  • the carrier tablet is coated with a carrier tablet film coat, e.g. to a 2-6% weight gain, for example using a coating not soluble in water (or not soluble in methanol or ethanol), for example using ethylcellulose (e.g. SureleaseTM) or methacrylic acid copolymer (e.g. EudragitTM) as the carrier tablet film coat.
  • a carrier tablet film coat e.g. to a 2-6% weight gain, for example using a coating not soluble in water (or not soluble in methanol or ethanol), for example using ethylcellulose (e.g. SureleaseTM) or methacrylic acid copolymer (e.g. EudragitTM) as the carrier tablet film coat.
  • the film covering the carrier tablet and comprising the compound or salt of the invention and the optional stabiliser is typically outside of and/or coated onto the carrier tablet film coat.
  • the carrier tablet can in particular have one or more recesses or depressions.
  • the film (which at least partially, e.g. only partially, covers the carrier tablet and which comprises the compound or salt of the invention and the optional stabiliser) is substantially present within the one or more recesses or depressions of the carrier tablet.
  • the above-mentioned dosage form (e.g. comprising a carrier tablet at least partially covered by a film comprising the compound or salt of the invention and an optional stabiliser) is further coated with an outer film coating.
  • the invention provides a method for preparing the above-mentioned pharmaceutical dosage form (comprising a carrier tablet at least partially covered by a film comprising the compound or salt of the invention and an optional stabiliser), wherein the method comprises dispensing a solution or suspension of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof and a stabiliser (e.g. citric acid or a salt thereof, e.g. present at about 2-3% w/v) onto a carrier tablet.
  • a stabiliser e.g. citric acid or a salt thereof, e.g. present at about 2-3% w/v
  • any solvent may be used provided that the stabiliser and any other excipients present in the film (which is to at least partially cover the carrier tablet) are soluble in the solvent.
  • the solvent is typically volatile.
  • the solvent should be pharmaceutically acceptable in any (residual) quantities in which it appears in the finished dosage form.
  • the solvent used in the method can include water, and/or an organic solvent such as methanol, ethanol, acetone, acetic acid and/or dichloromethane.
  • a mixture of solvents e.g. water-ethanol
  • the solvent is methanol.
  • the carrier tablet and the dispensed solution or suspension may be heated (e.g. in a forced air oven) to evaporate excess liquid and may result in the formation of a film upon at least a part of the surface of the carrier tablet.
  • the dosage form may then optionally be film coated, e.g. according to known methods, to create an outer film coating.
  • the carrier tablet used in the method for preparing the dosage form may have a recess or depression that provides a basin for the solution or suspension of the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the pharmaceutically acceptable salt thereof and the stabiliser to land after being dispensed.
  • biconcave tablets having recesses on two faces of the tablet are employed.
  • the above-mentioned dosage form comprising a carrier tablet of the present invention is produced by an apparatus described in WO 2005/123569, and more particularly is produced by an apparatus containing a dispensing module for accurately dispensing a predetermined amount of the solution or suspension of the 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or the pharmaceutically acceptable salt thereof and the stabiliser onto the carrier tablets.
  • the apparatus may also have a holding member for holding the carrier tablets, which may move continually along the apparatus as the dispensing module dispenses the solution or suspension onto each of the carrier tablets.
  • the apparatus may also have a drying system that dries or evaporates solvent from the solution or suspension deposited on each of the carrier tablets.
  • the holding member may move continually along the apparatus as the drying system dries the dosage on each of the carrier tablets.
  • the drying system may dry the dosage form by use of heated air, or by infrared or microwave heating.
  • the apparatus may also have a coating system that applies an outer film coating over the dosage form.
  • 1-(1-Methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof may be used in combination with other therapeutic agents.
  • 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof is intended for use in the treatment or prophylaxis (in particular treatment) of Alzheimer's disease, dementia, mild cognitive impairment, or a related neurodegenerative disorder, in particular in the treatment or prophylaxis (in particular treatment) of cognitive impairment(s) in Alzheimer's disease, dementia (e.g.
  • Lewy body dementia or vascular dementia mild cognitive impairment
  • a related neurodegenerative disorder e.g. in a mammal such as a human
  • medicaments claimed to be useful as either disease modifying or symptomatic treatments of Alzheimer's disease, dementia, mild cognitive impairment, or a related neurodegenerative disorder may be symptomatic agents, for example those known to modify cholinergic transmission such as M1 muscarinic receptor agonists or allosteric modulators, M2 muscarinic antagonists, acetylcholinesterase inhibitors (such as tetrahydroaminoacridine, donepezil e.g.
  • antipsychotics including typical antipsychotics (for example chlorpromazine, thioridazine, mesoridazine, fluphenazine, perphenazine, prochlorperazine, trifluoperazine, thiothixine, haloperidol, molindone or loxapine), atypical antipsychotics (for example clozapine, olanzapine, risperidone, quetiapine, ziprasidone, amisulpride or aripiprazole), glycine transporter 1 inhibitors and metabotropic receptor ligands; ii) drugs for extrapyramidal side effects,
  • the compounds may be administered either sequentially or simultaneously by any convenient route.
  • the invention thus provides, in a further aspect, a combination comprising 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine or a pharmaceutically acceptable salt thereof together with a further therapeutic agent or agents.
  • compositions comprising a combination as defined above together with a pharmaceutically acceptable carrier or excipient comprise a further aspect of the invention.
  • the individual components of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations.
  • the dose of each compound may differ from that when the compound is used alone.
  • the crude residue was purified by silica gel chromatography eluting with a gradient of from 0% to 30% ethyl acetate in hexane to yield the title compound as a pale yellow oil (2.19 g) (contaminated with trace amount of diisopropyl azodicarboxylate residue).
  • the reaction is cooled to room temperature, the methanol is evaporated off, the aqueous mixture is washed with dichloromethane (3 ⁇ 10 ml) and then is acidified to pH 2 with concentrated hydrochloric acid, and then the product is filtered off directly.
  • the batch is then seeded with pre-prepared triphenylphosphine oxide-diisopropyl hydrazinedicarboxylate adduct, and then allowed to stir for a further 1 hour before filtering.
  • the wet cake is washed with toluene (2 ⁇ 1 vol), and the combined mother liquors are transferred into a clean vessel.
  • the toluene solution is washed with 2M sodium hydroxide solution (5 vol) at 0-5° C., and then 3M sodium hydroxide solution (5 vol) is added and the reaction is heated to 80° C. The reaction is stirred for at least 2.5 hours, until HPLC shows no starting material.
  • the mixture is then cooled to 50° C. and toluene (5 vol) and water (5 vol) are added.
  • the layers are allowed to separate, and the aqueous layer is washed with toluene (10 vol) and then acidified to pH1 with 2.5M HCl solution (7.5 vol). The resultant slurry is filtered and the wet cake is washed with water (2 ⁇ 2 vol). The title product is dried at about 50° C. in a vacuum oven with a nitrogen bleed to constant probe temperature.
  • the filter cake in step 37, can be washed with toluene, instead of water, before the 50-75° C. vacuum drying of step 38.
  • the acid chloride product in dichloromethane was added to a stirred mixture of diethylaminomethylpolystyrene (844 mg, 3.2 mmol/g, 2.7 mmol, about 3 mole equivalents) and 1-isopropyl piperazine (115 mg, 0.90 mmol, e.g. available from Aldrich) in dichloromethane (10 ml) at room temperature. After 30 minutes, the mixture was loaded directly onto a silica flash column and eluted with a gradient of from 2% to 6% methanol (containing 10% 0.88 ammonia) in dichloromethane. The product-containing fractions were evaporated.
  • the reaction mixture was stirred at room temperature for 1.5 hours and was then quenched by the addition of a saturated aqueous solution of sodium hydrogen carbonate (500 ml) and ethyl acetate (1 litre).
  • the aqueous layer was extracted with ethyl acetate (400 ml) and the combined organic extracts were washed with water (2 ⁇ 400 ml).
  • the solvent was removed in vacuo and the residue was taken up in dichloromethane (150 ml), followed by the addition of a 1.0 M HCl solution in diethyl ether (200 ml).
  • the resultant white solid was then separated by filtration and was washed with dichloromethane to give the corresponding hydrochloride salt.
  • the mixture is cooled to 20° C. and any insoluble matter (e.g. any inorganics carried over in the 4-(tetrahydro-2H-pyran-4-yloxy)benzoic acid) is removed by filtration.
  • the clarified solution is transferred to a 10 L vessel in portions and is concentrated by distillation under reduced pressure to approximately 3 volumes to remove acetonitrile (e.g. using 50° C. jacket temperature and 200 mbar pressure reducing to 100 mbar pressure).
  • propan-2-ol (6 volumes) is added and the solution is concentrated further by distillation under reduced pressure to 5 volumes. After further propan-2-ol (8 volumes) is added, the solution is heated to 70° C.
  • Example 2 From analysis, the crystalline Form 1 of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride produced by Example 2 is believed not to be an entirely pure crystal form, and is believed to contain very approximately 20% of crystalline Form 2. Analyses of the crystalline Form 1 product produced by Example 2 include the following:
  • XRPD data were acquired on a PANalytical X'Pert Pro powder diffractometer, equipped with an X'Celerator detector.
  • the acquisition conditions were: radiation: Cu K ⁇ (copper K-alpha), generator tension: 40 kV, generator current: 45 mA, start angle: 2.0° 2 ⁇ (two-theta), end angle: 40.0° 2 ⁇ , step size: 0.0167° 2 ⁇ (two-theta).
  • the time per step was 31.750 s.
  • the sample was prepared by mounting a few milligrams of sample on a Si wafer (zero background) plates, resulting in a thin layer of powder.
  • the XRPD spectrum for crystalline Form 1 as prepared by Example 2 is shown in FIG. 1 .
  • An XRPD overlay spectrum is shown in FIG. 3 , comparing the XRPD peaks of crystalline Form 1 from Example 2 (top) to those of crystalline Form 2 from Example 3 (bottom), for comparison purposes.
  • the crystalline Form 1 XRPD peaks at 15.7° 2 ⁇ and 25.5° 2 ⁇ appear to be characteristic for crystalline Form 1 in that these peaks do not appear to be present in the XRPD spectrum of crystalline Form 2.
  • FT-IR spectrum was acquired over 64 scans at 4 cm ⁇ 1 resolution using a Nicolet Avatar 360 FT-IR spectrometer, fitted with a Diamond/ZnSe ATR Accessory.
  • the FT-IR spectrum for crystalline Form 1 as prepared by Example 2 is shown in FIGS. 4 and 5 , showing the spectral regions from 4000 to 675 cm ⁇ 1 and from 2000 to 675 cm ⁇ 1 respectively.
  • a solid-state NMR spectrum was obtained at a frequency of 90.55 MHz for 13 C observation using a 4-mm Bruker HFX MAS (magic-angle spinning) probe at a temperature of 296K, and a spinning speed of 8 kHz. Data were acquired using a cross polarisation sequence with side-band suppression. Several scans were acquired, with a relaxation delay of 10 seconds.
  • N-isopropylpiperazine (1-isopropylpiperazine, 0.66 wt, 13.33 g, obtained from Fluorochem ACI) was added to the reaction mixture, and heating was continued for 2 hours.
  • the mixture was concentrated to approx 2.5 volumes (50 mL) via vacuum distillation to give a thick mobile oil.
  • Propan-2-ol (240 mL, 12 volumes) was then added, and the mixture was concentrated by distillation under reduced pressure (100 mbar) to remove 2 volumes (40 mL).
  • the mixture was heated to 70° C. with stirring. 5-6N hydrochloric acid in Propan-2-ol (20 mL, 1 volume) was added to the mixture over 10 min. No crystallisation occurred until all of the acid had been added.
  • the reaction mixture was maintained at 70° C. overnight.
  • the mixture was cooled and the slurry was filtered.
  • the filtered solid was dried under air suction on the filter over 1 to 1.5 hours.
  • the title product was obtained as a
  • XRPD data were acquired on a PANalytical X'Pert Pro powder diffractometer, equipped with an X'Celerator detector.
  • the acquisition conditions were: radiation: Cu K ⁇ (copper K-alpha), generator tension: 40 kV, generator current: 45 mA, start angle: 2.0° 2 ⁇ (two-theta), end angle: 40.0° 2 ⁇ , step size: 0.0167° 2 ⁇ (two-theta).
  • the time per step was 31.750 s.
  • the sample was prepared by mounting a few milligrams of sample on a Si wafer (zero background) plates, resulting in a thin layer of powder.
  • the XRPD spectrum for crystalline Form 2 as prepared by Example 3 is shown in FIG. 2 .
  • An XRPD overlay spectrum is shown in FIG. 3 , comparing the XRPD peaks of crystalline Form 2 from Example 3 (bottom) to those of crystalline Form 1 from Example 2 (top), for comparison purposes.
  • the crystalline Form 2 XRPD peaks at 20.0° 2 ⁇ and 24.65° (or 24.7°) 2 ⁇ appear to be characteristic for crystalline Form 2 in that these peaks do not appear to be significantly present in the XRPD spectrum of crystalline Form 1.
  • FT-IR spectrum was acquired over 64 scans at 4 cm ⁇ 1 resolution using a Nicolet Avatar 360 FT-IR spectrometer, fitted with a Diamond/ZnSe ATR Accessory.
  • the FT-IR spectrum for crystalline Form 2 as prepared by Example 3 is shown in FIGS. 6 and 7 , showing the spectral regions from 4000 to 675 cm ⁇ 1 and from 2000 to 675 cm ⁇ 1 respectively.
  • An FT-IR overlay spectrum comparing these Form 2 peaks from Example 3 to those of crystalline Form 1 from Example 2 is shown in FIG. 8 , for comparison purposes, showing the spectral regions from 2000 to 675 cm ⁇ 1 .
  • a solid-state NMR spectrum was obtained at a frequency of 90.55 MHz for 13 C observation using a 4-mm Bruker HFX MAS (magic-angle spinning) probe at a temperature of 296K, and a spinning speed of 8 kHz. Data were acquired using a cross polarisation sequence with side-band suppression. Several scans were acquired, with a relaxation delay of 10 seconds.
  • Reaction progress is monitored by HPLC: the sample is prepared by quenching a drop of the reaction mixture into 1 mL of 5% butylamine solution in acetonitrile; this allows determination of residual 4-(tetrahydro-2H-pyran-4-yloxy)benzoic acid by derivatisation of the activated acid-imidazolide to the corresponding butylamide. This is generally recorded for information only since slurry inhomogeneity can give rise to inconsistent results (typically ⁇ 2% residual 4-(tetrahydro-2H-pyran-4-yloxy)benzoic acid is satisfactory at this stage).
  • 1-isopropylpiperazine (0.667 wt, 6.67 g, 1.15 mole equivalents) is added in one portion at about 65° C., followed by a line wash with acetonitrile (0.5 vol, 5 mL).
  • acetonitrile 0.5 vol, 5 mL
  • the resulting hazy solution is kept stirred at about 65° C. for at least ca. 2 hours (e.g. ca. 2-2.5 hours) before being sampled. Reaction progress is monitored by HPLC, using the method stated above.
  • the reaction is then allowed to cool and insoluble matter is removed by filtration.
  • the clarified solution is then concentrated by vacuum distillation to 2.5 to 3 volumes and 5% water in isopropanol (5 volumes, 50 mL) is added at ambient temperature.
  • the solution is then heated to about 65° C. and 5-6N HCl in isopropanol (0.9 volumes) is added in one charge. Crystallisation can commence shortly after the addition.
  • the resulting slurry is aged at 65° C. for 1.5 hours.
  • the slurry is then cooled to 55° C. over ca. 20 min and kept at 55° C. for 1.5 hours, is then cooled to 45° C. over ca. 20 min and kept at 45° C. for 1.5 hours, and is then allowed to cool to ambient temperature and the solid is filtered off (1 hour in total); the total cooling time is therefore about 4.5 to 4.75 hours.
  • a seed of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride (0.01 wt) is added and the resulting suspension is stirred at 58 ⁇ 3° C. for 30 min.
  • the suspension is then cooled to 0 ⁇ 3° C. over 2 hours before being aged at this temperature for 1 hour.
  • the solid is then filtered off under vacuum and washed with cold ethanol (3 vol).
  • the product is the dried in vacuo at 40° C. until constant probe temperature.
  • a membrane preparation containing histamine H3 receptors may be prepared in accordance with the following procedures:
  • DNA encoding the human histamine H3 gene was cloned into a holding vector, pcDNA3.1 TOPO (InVitrogen) and its cDNA was isolated from this vector by restriction digestion of plasmid DNA with the enzymes BamHI and Not-1 and ligated into the inducible expression vector pGene (InVitrogen) digested with the same enzymes.
  • the GeneSwitchTM system (a system where in transgene expression is switched off in the absence of an inducer and switched on in the presence of an inducer) was performed as described in U.S. Pat. Nos.
  • Ligated DNA was transformed into competent DH5 ⁇ E. coli host bacterial cells and plated onto Luria Broth (LB) agar containing ZeocinTM (an antibiotic which allows the selection of cells expressing the sh ble gene which is present on pGene and pSwitch) at 50 ⁇ g ml ⁇ 1 . Colonies containing the re-ligated plasmid were identified by restriction analysis. DNA for transfection into mammalian cells was prepared from 250 ml cultures of the host bacterium containing the pGeneH3 plasmid and isolated using a DNA preparation kit (Qiagen Midi-Prep) as per manufacturers guidelines (Qiagen).
  • CHO K1 cells previously transfected with the pSwitch regulatory plasmid (InVitrogen) were seeded at 2 ⁇ 10e6 cells per T75 flask in Complete Medium, containing Hams F12 (GIBCOBRL, Life Technologies) medium supplemented with 10% v/v dialysed foetal bovine serum, L-glutamine, and hygromycin (100 ⁇ g ml ⁇ 1 ), 24 hours prior to use. Plasmid DNA was transfected into the cells using Lipofectamine plus according to the manufacturers guidelines (InVitrogen). 48 hours post transfection cells were placed into complete medium supplemented with 500 ⁇ g ml ⁇ 1 ZeocinTM.
  • nM Mifepristone 10-14 days post selection 10 nM Mifepristone (InVitrogen), was added to the culture medium to induce the expression of the receptor. 18 hours post induction cells were detached from the flask using ethylenediamine tetra-acetic acid (EDTA; 1:5000; InVitrogen), following several washes with phosphate buffered saline pH 7.4 and resuspended in Sorting Medium containing Minimum Essential Medium (MEM), without phenol red, and supplemented with Earles salts and 3% Foetal Clone II (Hyclone).
  • EDTA ethylenediamine tetra-acetic acid
  • Positively stained cells were sorted as single cells into 96-well plates, containing Complete Medium containing 500 ⁇ g ml ⁇ 1 ZeocinTM and allowed to expand before reanalysis for receptor expression via antibody and ligand binding studies.
  • the cell pellet is resuspended in 10 volumes of homogenisation buffer (50 mM N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid (HEPES), 1 mM ethylenediamine tetra-acetic acid (EDTA), pH 7.4 with KOH containing 10e-4M leupeptin (acetyl-leucyl-leucyl-arginal; Sigma L2884) and 25 g/ml bacitracin (Sigma B0125)) supplemented with 1 mM phenylmethylsulfonyl fluoride (PMSF) and 2 ⁇ 10e-6M pepstain A (Sigma).
  • HEPES N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid
  • EDTA mM ethylenediamine tetra-acetic acid
  • pH 7.4 pH 7.4
  • KOH containing 10e-4M leupeptin acetyl
  • the cells are then homogenised by 2 ⁇ 15 second bursts in a 1 litre glass Waring blender, followed by centrifugation at 500 g for 20 minutes. The supernatant is then spun at 48,000 g for 30 minutes. The pellet is resuspended in homogenisation buffer (4 ⁇ the volume of the original cell pellet) by vortexing for 5 seconds, and then being forced by syringe through a 0.6 mm internal diameter needle. At this point the preparation is aliquoted into polypropylene tubes and stored at ⁇ 80° C.
  • the human H1 receptor was cloned generally using known procedures described in the literature [Biochem. Biophys. Res. Commun. 1994, 201(2), 894]. Chinese hamster ovary cells stably expressing the human H1 receptor were generated generally according to known procedures described in the literature [Br. J. Pharmacol. 1996, 117(6), 1071].
  • the compound of the invention or a pharmaceutically acceptable salt thereof may be tested for in vitro biological activity in accordance with the following assays:
  • the plate is sealed and after 2-6 hours, the plate is centrifuged for 5 min at 1500 rpm and counted on a Viewlux counter using a 613/55 filter for 5 min/plate. Data is analysed using a 4-parameter logistical equation. Basal activity used as minimum, i.e. wherein the histamine H3 antagonist iodophenpropit (30 uM, 0.5 ul) has been added to the well.
  • Adherent Chinese Hamster Ovary (CHO) cells stably expressing the recombinant human H 1 receptor were maintained in culture at 37° C. under 5% CO 2 in Alpha Minimum Essential Medium without ribonucleosides (Gibco Invitrogen) supplemented with 10% dialysed foetal calf serum and 200 mM Glutamine. These cells, expressing the human H1 receptor, were snap frozen and stored ready for assay.
  • HBSS medium (CaCl 2 .2H 2 O 1.26 mM, Glucose 5.55 mM, KCl 5.36 mM, MgSO 4 (anhyd.) 0.81 mM, NaCl 136.89 mM, KH 2 PO 4 (anhyd.) 0.41 mM, HEPES 20 mM, NaHCO 3 4.16 mM) containing the cytoplasmic calcium indicator, Fluo-4 in the acetylmethyl form (4 mM), 2.5 mM Probenecid and 250 uM Brilliant Black (Molecular Devices) at 37° C. for 60 min.
  • HBSS medium CaCl 2 .2H 2 O 1.26 mM, Glucose 5.55 mM, KCl 5.36 mM, MgSO 4 (anhyd.) 0.81 mM, NaCl 136.89 mM, KH 2 PO 4 (anhyd.) 0.41 mM, HEPES 20
  • the loaded cells were then incubated with test compound for 30 min at 37° C.
  • the plates were then placed into a FLIPR (Molecular Devices, UK) for testing in antagonist mode, where a pre-determined concentration of Histamine (approximately 4 ⁇ EC50) was added while cell fluorescence ( ⁇ ex 488 nm, ⁇ em 540 nm) was monitored.
  • Functional antagonism is indicated by a suppression of histamine induced increase in fluorescence, as measured by the FLIPRTM system (Molecular Devices). By means of concentration effect curves, functional affinities are determined using standard pharmacological mathematical analysis.
  • 1-(1-Methyl ethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride (e.g. Example E1) was tested in the histamine H1 functional antagonist assay or a similar H1 functional antagonist assay. Again, the result is expressed as a functional pK i (fpK i ) value and is an average of a number of experiments.
  • 1-(1-Methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride (e.g. Example E1) exhibited antagonism approximately ⁇ 5.6 fpK i .
  • the tissues were rapidly thawed and homogenised in approximately 30 volumes of ice cold assay reaction buffer.
  • the assay reaction buffer contained 50 mM Tris-HCl (made up using Trizma pre-set crystals pH 7.7 @25° C., Sigma cat. No. T8068-250G) and 5 mM EDTA, with a final buffer pH of 7.2 to 7.8, usually about 7.4.
  • the crude homogenate 600-800 ⁇ g/well was then used to measure H3 receptor binding using [ 3 H]-R- ⁇ -methylhistamine as radioligand.
  • Assays measuring total binding of [ 3 H]-R- ⁇ -methylhistamine consisted of 50 ⁇ l assay reaction buffer, 400 ⁇ l of homogenate (corresponding to 600-800 ⁇ g/well) and 50 ⁇ l of 2 nM R( ⁇ ) ⁇ -Methyl[imidazole-2,5(n)-3H]histamine dihydrochloride ([ 3 H]-R- ⁇ -methylhistamine; specific activity, 24 Ci mmol ⁇ 1 , Amersham Biosciences, catalogue no. TRK1017). Incubations with [ 3 H]-R- ⁇ -methylhistamine were for 45 min at 30° C.
  • Non-specific binding of [ 3 H]-R- ⁇ -methylhistamine was determined in parallel using the same assay except that 50 ⁇ l of 10 ⁇ M imetit (an H3 receptor agonist, e.g. available from Tocris) was used instead of the 50 ⁇ l of assay reaction buffer.
  • the experiments were terminated by rapid filtration through Whatman GF/B filters (pre-soaked in 0.3% v/v polyethyleneimine (PEI)), and then the filters were washed through with 4 ⁇ 5 ml of ice cold harvesting buffer.
  • the harvesting buffer contained 50 mM Tris-HCl (from Trizma pre-set crystals pH 7.7 @ 25° C.) and 5 mM MgCl 2 , with a final buffer pH of 7.2 to 7.8, usually about 7.4. Filters were dried and added to vials each containing 4 ml Ultima Gold MV scintillation fluid (Hewlett Packard) and radioactivity determined by liquid scintillation spectrometry using a Packard Tri-Carb 2500TR liquid scintillation counter.
  • Tris-HCl from Trizma pre-set crystals pH 7.7 @ 25° C.
  • MgCl 2 final buffer pH of 7.2 to 7.8
  • Filters were dried and added to vials each containing 4 ml Ultima Gold MV scintillation fluid (Hewlett Packard) and radioactivity determined by liquid scintillation spectrometry using a Packard Tri-Carb 2500TR liquid scintillation counter.
  • Protein Assay Dye Reagent Concentrate catalogue no. 500-0006; from Bio-Rad Laboratories GmbH, Heidemannstrasse 164, 80939 Muenchen, Germany; or from Bio-Rad, York, UK
  • bovine serum albumin bovine serum albumin
  • FIGS. 10 , 11 , 12 , 13 and 14 These studies, and results arising therefrom, are illustrated in part by the attached FIGS. 10 , 11 , 12 , 13 and 14 , already briefly described.
  • PET is a nuclear imaging technique that enables the measurement of the four-dimensional (three space, one time) distribution of a radiopharmaceutical in the living body.
  • a bioactive molecule (which binds to the receptor of interest, in the present case the histamine H3 receptor) is modified by exchanging one of its atoms by a positron emitting nuclei (e.g. 15 O, 11 C, 18 F, etc).
  • the radioactive molecule (radiopharmaceutical) is then intravenously injected into the subject.
  • a positron-emitting atom undergoes radioactive decay by releasing a positron from its nucleus.
  • the positron loses kinetic energy and interacts with an electron by annihilation.
  • the annihilation results in two high energy photons (2 ⁇ 511 keV) emitted at 180° to each other.
  • the high energy photon-pair generated by the positron-electron annihilation, emitted at 180°, can be detected externally.
  • a ring of crystal detectors in the PET scanner senses the presence of two photons generated simultaneously and records data for the two detectors pair that sensed the two photons, enabling localization of the activity. This is generally done millions of times during the course of a PET scan. 3D images are sub-sequentially generated using tomographic reconstruction techniques.
  • SA specific activity
  • Receptor occupancy of a non-radiolabelled test compound can be measured indirectly by measuring the reduction in specific binding of the radioligand as a consequence of competitive binding ( FIG. 10 ).
  • the occupancy of the drug candidate can be measured indirectly by the reduction in radioligand specific binding to receptors.
  • B A is the concentration of available receptors sites. Notice how B A changes between baseline, and 10 min, 2.5 hours, and 6 hours after administration of the test compound, as a consequence of the presence of different concentrations of the drug candidate in tissue.
  • a baseline scan is performed where a small mass of the radioligand (in the ⁇ g range such that the self-occupancy of the radioligand is minimal, ⁇ 10%) is administered to the subject.
  • BP ND the binding potential
  • the unlabeled drug candidate is administered to the subject and sub-sequent scans are acquired at time points of interest and binding potentials are estimated.
  • the drug candidate occupancy at different time points is calculated as the percentage change of the binding potential measurements with respect to baseline (J. Passchier, A. Gee, A. Willemsen, W. Vaalburg, and A. van Waarde, “Measuring drug-related receptor occupancy with positron emission tomography,” Methods, 2002, vol. 27, pp. 278-286).
  • [ 11 C]GSK189254 is [ 11 C-N-methyl]-6-(3-cyclobutyl-2,3,4,5-tetrahydro-1H-benzo[d]azepin-7-yloxy)-nicotinamide, the structure of which is
  • the radioligand [ 11 C]GSK189254 was synthesized immediately before each PET scan.
  • the crude [ 11 C]GSK189254-containing reaction product was subject to HPLC purification performed using a reverse phase C 18 column (Waters, X-Terra® RP18, 19 ⁇ 100 mm, 5 mm) at 10 mL/min flow rate with a mobile phase consisting of 17% of acetonitrile in a 0.1 N aqueous buffer solution of ammonium formate at pH 4.
  • the [ 11 C]GSK189254-containing product fraction collected was concentrated in vacuo to remove the acetonitrile and reformulated in 0.9% aqueous sodium chloride solution.
  • the radioligand [ 11 C]GSK189254 in the above-mentioned vehicle which was effectively a mixture of aqueous ammonium formate buffer and saline, was administered intravenously (i.v.) over about 1 minute as a bolus.
  • the amount of radioactivity injected into each pig via [ 11 C]GSK189254 was generally about 250 to about 400 MBq (ideally about 300 MBq).
  • the volume of [ 11 C]GSK189254+vehicle injected per pig depends on the yield obtained from the last HPLC purification, but generally ranged from about 2 to about 11 ml.
  • salt A 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride
  • PET images were aligned to a stereotaxic atlas and regional time activity curves were obtained for the target regions of interest (frontal cortex, hippocampus, putamen, caudate, diencephalon, medial thalamus, lateral thalamus, vermis, pons, mesencephalon, and medulla oblongata).
  • the simplified reference tissue model (SRTM) (A. A. Lammertsma and S. P. Hume, “Simplified reference tissue model for PET receptor studies,” Neuroimage, 1996, vol. 4, pp. 153-158) was fitted to each regional time activity curve using the cerebellum as the reference region:
  • C t (t) is the activity concentration in the target region
  • C r (t) is the activity concentration in the reference region (cerebellum)
  • R 1 is the ratio of influx (K 1 ) between the target and reference region
  • k 2 is the tissue-plasma efflux rate constant in the target region
  • BP ND is the binding potential of the target region.
  • the binding potential can be defined as
  • ⁇ ND is the radioligand free fraction in tissue
  • B A is the available concentration of binding sites
  • K d is the equilibrium dissociation constant of the radioligand-receptor complex
  • Receptor occupancy can be calculated as the percentage change in BP ND between the baseline and post drug scan (J. Passchier, A. Gee, A. Willemsen, W. Vaalburg, and A. van Waarde, “Measuring drug-related receptor occupancy with positron emission tomography,” Methods, 2002, vol. 27, pp. 278-286):
  • Occ corrected Occ ⁇ ( 1 + BP ND ref 1 + B ND ref ⁇ Occ )
  • the temporal occupancy profile at 10, 150, and 360 minutes (Occ(t)) post administration of the drug candidate and the plasma concentration of the drug candidate (C p (t)) measured throughout the PET whole scan were used to derive PK/RO (pharmacokinetic/receptor occupancy) model parameter estimates (f p , k on and k off ) from an indirect model (k on -k off limited model)
  • PK/RO pharmacokinetic/receptor occupancy
  • k on and k off are respectively the rate constants defining the speed that the test compound attaches or de-attaches to or from the H3 receptor
  • f p is the protein-free fraction in plasma of the test compound (here, salt A or salt B).
  • FIG. 12 graph A, shows the average (mean) plasma concentration over time of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride (“salt A”, within the present invention, filled circles) and 1-(isopropyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ hexahydro-1H-1,4-diazepine hydrochloride (“salt B”, a comparator compound, filled diamonds).
  • FIG. 12 graph B, shows the average (mean) measured H3 receptor occupancy time course at three time points; and the k on -k off limited model fitted to it for “salt A” within the present invention (measurements as filled circles, and model fit as solid line), and for “salt B” a comparator compound (measurements as filled diamonds, and model fit as dashed line).
  • FIG. 13 parts A and B, are graphs showing the data from FIG. 12 for salt A alone such as to show the average (mean) plasma concentration over time and average (mean) H3 receptor occupancy time course respectively for “salt A”.
  • FIG. 13 , parts C and D are graphs showing the data from FIG. 12 for salt B alone such as to show the average (mean) plasma concentration over time and average (mean) H3 receptor occupancy time course respectively for “salt B”.
  • Table 1 shows the estimated parameters k on , k off , K d (k off /k on ), and the plasma clearance for each individual study (i.e. for each individual pig).
  • the last row in Table 1 (“average model”) shows the estimated parameters derived from the average plasma data and average occupancy ( FIGS. 12 , 13 ); it can be noticed that these “average model” parameters appear to be in general agreement with the average parameters (“mean”) estimated individually for each scan (second row from the bottom).
  • Table 2 shows the measured H3 receptor occupancy at three time points, in each the three individual studies performed (in each of the three individual pigs studied), for each test compound, salt A and salt B.
  • Toxicity Study 1 Toxicity of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine Hydrochloride in a 7-Day, Oral Repeat-Dose Study in Male Sprague Dawley Rats
  • the objective of this study was to determine the toxicity of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride, and the toxicokinetics of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]-carbonyl ⁇ piperazine hydrochloride (measured as the free base), in a 7-day, oral repeat-dose study in male Sprague Dawley rats.
  • 1-(1-Methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride was formulated as a suspension in 1% (w/v) aqueous methylcellulose and administered to male rats (four per group), at doses of 0 (vehicle, control), 30, 100 or 300 mg/kg/day for 7 days by oral gavage, at a dose volume of 10 mL/kg. Three male rats were added at each non-zero dose level for toxicokinetic evaluation. All doses and concentrations, including analyte concentration in plasma, are expressed in terms of the parent “free base” compound.
  • the following endpoints/parameters were evaluated for toxicology animals: clinical observations, body weights, food consumption measurements, selected haematology and selected clinical chemistry results, liver weights, selected macroscopic and microscopic observations, and selected hepatic gene expression analysis.
  • Toxicokinetic evaluation was performed on samples collected from satellite animals on Days 1 and 7. For male rats dosed at 30 or 100 mg/kg/day, histopathology observations carried out were on kidneys, mesentery and mandibular lymph node only.
  • Toxicity Study 2 Toxicity of 1-(1-methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine Hydrochloride in a 7-Day, Oral, Dose Range-Finding Study in Sprague Dawley Rats
  • Glandular dilation of the fundic region of the stomach was observed in most female rats given doses of 300 mg/kg/day. This change was also present in two controls (one male and one female) and its significance is unclear at this stage.
  • 1-(1-Methylethyl)-4- ⁇ [4-(tetrahydro-2H-pyran-4-yloxy)phenyl]carbonyl ⁇ piperazine hydrochloride appears to be well tolerated or moderately well tolerated in male and female Sprague Dawley rats after 7 days of oral-repeat-dosing at doses of up to 300 mg/kg/day (measured as the free base).

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Anesthesiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Pyrane Compounds (AREA)
US12/203,946 2007-09-06 2008-09-04 Piperazine derivative having affinity for the histamine h3 receptor Abandoned US20090170869A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GB0717336A GB0717336D0 (en) 2007-09-06 2007-09-06 Novel compound
GB0717336.2 2007-09-06
GB0814987.4 2008-08-15
GB0814987A GB0814987D0 (en) 2008-08-15 2008-08-15 Novel compound

Publications (1)

Publication Number Publication Date
US20090170869A1 true US20090170869A1 (en) 2009-07-02

Family

ID=39885024

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/676,158 Expired - Fee Related US8288389B2 (en) 2007-09-06 2008-09-04 Piperazine derivative having affinity for the histamine H3 receptor
US12/203,946 Abandoned US20090170869A1 (en) 2007-09-06 2008-09-04 Piperazine derivative having affinity for the histamine h3 receptor

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/676,158 Expired - Fee Related US8288389B2 (en) 2007-09-06 2008-09-04 Piperazine derivative having affinity for the histamine H3 receptor

Country Status (17)

Country Link
US (2) US8288389B2 (pt)
EP (1) EP2195307B1 (pt)
JP (1) JP5379796B2 (pt)
KR (1) KR20100059944A (pt)
CN (1) CN101848903A (pt)
AR (1) AR068370A1 (pt)
AT (1) ATE554081T1 (pt)
AU (1) AU2008294708B2 (pt)
BR (1) BRPI0816304A2 (pt)
CA (1) CA2697941A1 (pt)
CL (1) CL2008002622A1 (pt)
EA (1) EA201070338A1 (pt)
ES (1) ES2384414T3 (pt)
MX (1) MX2010002606A (pt)
PE (1) PE20091199A1 (pt)
TW (1) TW200918527A (pt)
WO (1) WO2009030716A1 (pt)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100204242A1 (en) * 2007-09-06 2010-08-12 Glaxo Group Limited Piperazine Derivative Having Affinity for the Histamine H3 Receptor
WO2012114348A1 (en) 2011-02-23 2012-08-30 Suven Life Sciences Limited Novel compounds as histamine h3 receptor ligands
WO2014030170A1 (en) 2012-08-23 2014-02-27 Suven Life Sciences Limited Acrylamide compounds as histamine h3 receptor ligands
US10183938B2 (en) 2014-12-16 2019-01-22 Axovant Sciences Gmbh Geminal substituted quinuclidine amide compounds as agonists of α-7 nicotonic acetylcholine receptors
US10370370B2 (en) 2015-06-10 2019-08-06 Axovant Sciences Gmbh Aminobenzisoxazole compounds as agonists of α7-nicotinic acetylcholine receptors
US10428062B2 (en) 2015-08-12 2019-10-01 Axovant Sciences Gmbh Geminal substituted aminobenzisoxazole compounds as agonists of α7-nicotinic acetylcholine receptors

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0324159D0 (en) 2003-10-15 2003-11-19 Glaxo Group Ltd Novel compounds
US9108948B2 (en) 2006-06-23 2015-08-18 Abbvie Inc. Cyclopropyl amine derivatives
CN103382174A (zh) 2006-06-23 2013-11-06 雅培制药有限公司 作为组胺h3受体调节物的环丙胺衍生物
JP2012500192A (ja) * 2008-08-15 2012-01-05 グラクソ グループ リミテッド 1−イソプロピル−4−{[4−(テトラヒドロ−2h−ピラン−4−イルオキシ)フェニル]カルボニル}ヘキサヒドロ−1h−1,4−ジアゼピンの塩およびその調製方法
US9186353B2 (en) 2009-04-27 2015-11-17 Abbvie Inc. Treatment of osteoarthritis pain
WO2012037258A1 (en) 2010-09-16 2012-03-22 Abbott Laboratories Processes for preparing 1,2-substituted cyclopropyl derivatives
WO2013151982A1 (en) 2012-04-03 2013-10-10 Arena Pharmaceuticals, Inc. Methods and compounds useful in treating pruritus, and methods for identifying such compounds
EP2647377A1 (en) 2012-04-06 2013-10-09 Sanofi Use of an h3 receptor antagonist for the treatment of alzheimer's disease
EP3378477A1 (en) * 2017-03-21 2018-09-26 BIOPROJET Pharma New therapeutical use of h3-ligands

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU6179998A (en) 1997-02-24 1998-09-09 Zymogenetics Inc. Calcitonin mimetics
US6268367B1 (en) 1998-02-23 2001-07-31 Zymogenetics, Inc. Piperazine derivatives for treating bone deficit conditions
NZ523526A (en) 2000-07-20 2004-10-29 Neurogen Corp Diaryl piperazines as capsaicin receptor ligands
EP1311482B1 (en) 2000-08-08 2007-02-21 Ortho-McNeil Pharmaceutical, Inc. Non-imidazole aryloxypiperidines as h3 receptor ligands
AU2002344951A1 (en) 2001-07-02 2003-01-21 Boehringer Ingelheim International Gmbh Substituted piperazine and diazepanes as histamine h3 receptor agonists
DE10144226A1 (de) 2001-09-07 2003-03-27 Henkel Kgaa Neue verbrückte p-Phenylendiamine
NZ531378A (en) 2001-09-19 2006-11-30 Aventis Pharma S Indolizines as kinase protein inhibitors suitable for treating solid tumours
AU2003203148A1 (en) 2002-02-05 2003-09-02 High Point Pharmaceuticals, Llc Novel aryl- and heteroarylpiperazines
GB0224084D0 (en) 2002-10-16 2002-11-27 Glaxo Group Ltd Novel compounds
AU2003274053A1 (en) 2002-10-22 2004-05-13 Glaxo Group Limited Aryloxyalkylamine derivates as h3 receptor ligands
BRPI0315644B8 (pt) * 2002-10-23 2021-05-25 Janssen Pharmaceutica Nv compostos de piperazinil e diazapanil benzamidas e benzotioamidas, suas composições e respectivos usos
ES2401216T3 (es) 2002-12-20 2013-04-17 Glaxo Group Limited Nuevos derivados de benzazepina
US7449464B2 (en) 2003-03-12 2008-11-11 Kudos Pharmaceuticals Limited Phthalazinone derivatives
PL1615909T3 (pl) 2003-04-23 2009-01-30 Glaxo Group Ltd Pochodne piperazyny i ich zastosowanie w leczeniu chorób neurologicznych i psychicznych
GB0324159D0 (en) * 2003-10-15 2003-11-19 Glaxo Group Ltd Novel compounds
PL1802307T3 (pl) 2004-10-15 2008-07-31 Glaxo Group Ltd Pochodne pirolidyny jako ligandy receptorów histaminowych
UA93892C2 (ru) 2005-10-31 2011-03-25 Янссен Фармацевтика H.B. Способ получения производных циклопропиламида (варианты
EA014905B1 (ru) 2005-10-31 2011-02-28 Янссен Фармацевтика Н.В. Новый способ получения пиперазинильных производных бензамида
NZ577603A (en) 2006-12-14 2012-04-27 Janssen Pharmaceutica Nv Process for the preparation of piperazinyl and diazepanyl benzamide derivatives
ES2442926T3 (es) 2007-08-22 2014-02-14 Astrazeneca Ab Derivados de ciclopropilamida `978
JP5379796B2 (ja) 2007-09-06 2013-12-25 グラクソ グループ リミテッド ヒスタミンh3受容体に対しアフィニティーを有するピペラジン誘導体
UA103468C2 (ru) 2007-11-20 2013-10-25 Янссен Фармацевтика Н.В. Соединения циклоалкилокси- и гетероциклоалкилоксипиридина как модуляторы гистаминового рецептора н3
US9186353B2 (en) * 2009-04-27 2015-11-17 Abbvie Inc. Treatment of osteoarthritis pain

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100204242A1 (en) * 2007-09-06 2010-08-12 Glaxo Group Limited Piperazine Derivative Having Affinity for the Histamine H3 Receptor
US8288389B2 (en) 2007-09-06 2012-10-16 Glaxo Group Limited Piperazine derivative having affinity for the histamine H3 receptor
WO2012114348A1 (en) 2011-02-23 2012-08-30 Suven Life Sciences Limited Novel compounds as histamine h3 receptor ligands
US9079888B2 (en) 2011-02-23 2015-07-14 Suven Life Sciences Limited Compounds as histamine H3 receptor ligands
WO2014030170A1 (en) 2012-08-23 2014-02-27 Suven Life Sciences Limited Acrylamide compounds as histamine h3 receptor ligands
US10183938B2 (en) 2014-12-16 2019-01-22 Axovant Sciences Gmbh Geminal substituted quinuclidine amide compounds as agonists of α-7 nicotonic acetylcholine receptors
US10370370B2 (en) 2015-06-10 2019-08-06 Axovant Sciences Gmbh Aminobenzisoxazole compounds as agonists of α7-nicotinic acetylcholine receptors
US10428062B2 (en) 2015-08-12 2019-10-01 Axovant Sciences Gmbh Geminal substituted aminobenzisoxazole compounds as agonists of α7-nicotinic acetylcholine receptors

Also Published As

Publication number Publication date
WO2009030716A1 (en) 2009-03-12
TW200918527A (en) 2009-05-01
JP2010538041A (ja) 2010-12-09
KR20100059944A (ko) 2010-06-04
BRPI0816304A2 (pt) 2015-03-17
JP5379796B2 (ja) 2013-12-25
EA201070338A1 (ru) 2010-10-29
MX2010002606A (es) 2010-03-30
US20100204242A1 (en) 2010-08-12
EP2195307B1 (en) 2012-04-18
CA2697941A1 (en) 2009-03-12
AU2008294708A1 (en) 2009-03-12
CL2008002622A1 (es) 2009-11-27
ES2384414T3 (es) 2012-07-04
AR068370A1 (es) 2009-11-11
US8288389B2 (en) 2012-10-16
AU2008294708B2 (en) 2012-03-15
ATE554081T1 (de) 2012-05-15
PE20091199A1 (es) 2009-09-12
EP2195307A1 (en) 2010-06-16
CN101848903A (zh) 2010-09-29

Similar Documents

Publication Publication Date Title
US8288389B2 (en) Piperazine derivative having affinity for the histamine H3 receptor
US10981895B2 (en) Crystalline forms and processes for the preparation of cannabinoid receptor modulators
AU2009236256B2 (en) Small molecule inhibitors of the pleckstrin homology domain and methods for using same
EP3937943A1 (en) Novel small molecule inhibitors of tead transcription factors
IL310682A (en) The history of benzo a heteroaromatic ring containing nitrogen and its use in medicine
WO2015055071A1 (zh) 稠合杂环化合物、其制备方法、药物组合物和用途
US20140080804A1 (en) Novel 6-arylamino pyridone carboxamide as mek inhibitors
JPWO2020045334A1 (ja) 光学活性なアザビシクロ環誘導体
JP2012516321A (ja) デルター5−デサチュラーゼ阻害薬
JPH037257A (ja) ピリジン誘導体及びそれを有効成分とする向精神剤
JP4225894B2 (ja) 縮合環式基を有する環状ジアミン化合物
WO2024000401A1 (en) 2-((1-(6-methyl-4-oxo-3,4-dihydroquinazolin-8-yl)ethyl)amino)benzoic acid derivatives and similar compounds as pi3k inhibitors for the treatment of e.g. cancer
WO2016041201A1 (zh) 噻吩并嘧啶类衍生物、其制备方法及其在医药上的应用
CN114105977B (zh) 雌激素受体调节剂化合物及其用途
JP6662864B2 (ja) ガンマ−ラクタムのエステルプロドラッグ及びそれらの使用
TW202246230A (zh) 醯胺衍生物及其應用
TW202210488A (zh) 吡嗪類衍生物及其在抑制shp2中的應用
JPH05331065A (ja) セロトニン神経系関連疾患治療剤
NZ614787B2 (en) Crystalline forms and processes for the preparation of condensed azacycles ( cannabinoid receptor modulators)
JPH09132579A (ja) ベンゾイルピペリジン誘導体およびその塩

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLAXO GROUP LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BEST, DESMOND JOHN;ORLEK, BARRY SIDNEY;RASSIAS, GERACIMOS;AND OTHERS;REEL/FRAME:023033/0601;SIGNING DATES FROM 20081010 TO 20081117

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION