US20020019396A1 - Pharmaceutically active morpholinol - Google Patents

Pharmaceutically active morpholinol Download PDF

Info

Publication number
US20020019396A1
US20020019396A1 US09/886,391 US88639101A US2002019396A1 US 20020019396 A1 US20020019396 A1 US 20020019396A1 US 88639101 A US88639101 A US 88639101A US 2002019396 A1 US2002019396 A1 US 2002019396A1
Authority
US
United States
Prior art keywords
formula
compound
morpholinol
chlorophenyl
uptake
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.)
Granted
Application number
US09/886,391
Other versions
US6391875B2 (en
Inventor
Phillip Morgan
David Musso
John Partridge
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.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB9801230.5A external-priority patent/GB9801230D0/en
Priority to US09/886,391 priority Critical patent/US6391875B2/en
Application filed by Individual filed Critical Individual
Publication of US20020019396A1 publication Critical patent/US20020019396A1/en
Priority to US10/147,588 priority patent/US6855820B2/en
Priority to US10/150,287 priority patent/US6734213B2/en
Priority to US10/150,341 priority patent/US6998400B2/en
Priority to US10/150,339 priority patent/US7098206B2/en
Publication of US6391875B2 publication Critical patent/US6391875B2/en
Application granted granted Critical
Priority to US10/944,814 priority patent/US20050096318A1/en
Priority to US11/407,192 priority patent/US20060189612A1/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • 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
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • 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
    • 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
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/34Tobacco-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents

Definitions

  • This invention relates to an optically pure morpholinol, pharmaceutical formulations containing it and processes for their preparation and use.
  • Bupropion hydrochloride ( ⁇ )-1-(3-chlorophenyl)-2-[(1,1-dimethylethyl)-amino]-1-propanone hydrochloride, is the active ingredient of Wellbutrin® which is marketed in the United States for the treatment of depression. It is also the active ingredient of Zyban® which is marketed in the United States as an aid to smoking cessation. Bupropion is a relatively weak inhibitor of the neuronal uptake of noradrenaline (NA), serotonin and dopamine (DA), and does not inhibit monoamine oxidase.
  • NA noradrenaline
  • DA dopamine
  • Bupropion is extensively metabolized in man as well as laboratory animals.
  • Urinary and plasma metabolites include biotransformation products formed via hydroxylation of the tert-butyl group and/or reduction of the carbonyl group of bupropion.
  • Four basic metabolites have been identified. They are the erythro- and threo-amino alcohols of bupropion, the erythro-amino diol of bupropion, and a morpholinol metabolite. These metabolites of bupropion are pharmacologically active, but their potency and toxicity relative to bupropion have not been fully characterized. Because the plasma concentrations of the metabolites are higher than those of bupropion, they may be of clinical importance.
  • the present invention provides, in one aspect, a compound of formula (I), (+)-(2S, 3S)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol or pharmaceutically acceptable salts and solvates thereof.
  • compositions comprising a compound of formula (I) or pharmaceutically acceptable salts and solvates thereof together with one or more pharmaceutically acceptable carriers, diluents or excipients.
  • a further aspect of the present invention is the use of a compound of formula (I) or pharmaceutically acceptable salts and solvates thereof in therapy.
  • Yet another aspect of the invention provides methods of treating depression, attention deficit hyperactivity disorder (ADHD), obesity, migraine, pain, sexual dysfunction, Parkinson's disease, Alzheimer's disease, or addiction to cocaine or tobacco products in a human or animal subject comprising the administration to said subject of an effective amount of a compound of formula (I) or pharmaceutically acceptable salts and solvates thereof or pharmaceutical compositions thereof.
  • ADHD attention deficit hyperactivity disorder
  • a compound of formula (I) or pharmaceutically acceptable salts and solvates thereof or pharmaceutical compositions thereof comprising the administration to said subject of an effective amount of a compound of formula (I) or pharmaceutically acceptable salts and solvates thereof or pharmaceutical compositions thereof.
  • Yet another aspect of the present invention is the use of the compound of formula (I) or pharmaceutically acceptable salts and solvates thereof or pharmaceutical compositions thereof in the preparation of a medicament for the treatment of depression, attention deficit hyperactivity disorder (ADHD), obesity, migraine, pain, sexual dysfunction, Parkinson's disease, Alzheimer's disease, addiction to cocaine or tobacco products.
  • ADHD attention deficit hyperactivity disorder
  • FIG. 1 Effect of Compounds at 25 mg/kg (ip) on TBZ-Induced Depression
  • the compound of formula (I) or pharmaceutically acceptable salts and solvates thereof may be prepared by first synthesizing the racemate of the morpholinol metabolite of bupropion and subsequently separating the (+) and ( ⁇ ) enantiomers of the racemate via HPLC.
  • racemate of the morpholinol metabolite of bupropion hydrochloride ((+/+)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride) may be synthesized by the following process.
  • (+/ ⁇ )-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol may be converted to (+/ ⁇ )-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride by the following process.
  • a 6.0 g sample was dissolved in diethyl ether, chilled in an ice bath and ethereal hydrogen chloride added until the mixture was acidic.
  • (+/ ⁇ )-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride may be converted back to its free base by the following process.
  • a 3.0 g sample of (+/ ⁇ )-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride was dissolved in water (100 mL) and diethyl ether was added (200 mL). The mixture was chilled in an ice bath and the pH was adjusted to >10 with 1.0 N aqueous sodium hydroxide.
  • (+) and ( ⁇ ) enantiomers of (+/ ⁇ )-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol may be separated by the following process.
  • (+/ ⁇ )-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol (2.54 gms.) was dissolved in 250 ml of 2:8 Isopropyl alcohol: Hexane (both HPLC grade).
  • a Daicel Chiralcel OD column (2 ⁇ 25 cm.) was equilibrated for one hour at 8 ml./min.
  • optical isomer 1 weighed 1.25 gm. and optical isomer 2 weighed 1.26 gm.
  • Hydrochloride salts of the separated enantiomers were obtained by the following processes. 1.25 g (0.005 mol) of optical isomer 1 (retention time 6.117 min) (( ⁇ )-(2R,3R)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol) was dissolved in diethyl ether. The solution was filtered and the filtrate was chilled in an ice-bath adding ethereal hydrogen chloride until the solution was acidic. After standing at ambient temperature for 24 h, the resulting solid was filtered, washed with diethyl ether and dried in a vacuum oven at 60° C.
  • the amount of compound of formula (1) required to achieve the desired therapeutic effect will, of course depend on a number of factors, for example, the mode of administration, the recipient and the condition being treated.
  • the daily dose will be in the range of 0.02 to 5.0 mg/kg. More particular ranges include 0.02 to 2.5 mg/kg, 0.02 to 1.0 mg/kg, 0.02 to 0.25 mg/kg, 0.02 to 0.15 mg/kg and 0.02 to 0.07 mg/kg.
  • the compound of formula (I) may be employed in the treatment of depression, attention deficit hyperactivity disorder (ADHD), obesity, migraine, pain, sexual dysfunction, Parkinson's disease, Alzheimer's disease, addiction to cocaine or tobacco products as the compound per se, but is preferably presented with one or more pharmaceutically acceptable carriers, diluents or excipients in the form of a pharmaceutical formulation.
  • the carriers, diluents and exipients must, of course, be acceptable in the sense of being compatible with the other ingredients of the formulation and must not be deleterious to the recipient.
  • the carrier may be a solid or a liquid, or both, and is preferably formulated with the agent as a unit-dose formulation, for example, a tablet.
  • the formulations include those suitable for oral, rectal, topical, buccal (e.g. sub-lingual) and parenteral (e.g. subcutaneous, intramuscular, intradermal or intravenous) administration.
  • buccal e.g. sub-lingual
  • parenteral e.g. subcutaneous, intramuscular, intradermal or intravenous
  • Formulations suitable for buccal (sub-lingual) administration include lozenges comprising a compound of formula (I) in a flavoured base, usually sucrose and acacia or tragacanth, and pastilles comprising the agent in an inert base such as gelatin and glycerin or sucrose and acacia.
  • Formulations of the present invention suitable for parenteral administration conveniently comprise sterile aqueous preparations of a compound of formula (I), preferably isotonic with the blood of the intended recipient. These preparations are preferably administered intravenously, although administration may also be effected by means of subcutaneous, intramuscular, or intradermal injection. Such preparations may conveniently be prepared by admixing the agent with water and rendering the resulting solution sterile and isotonic with the blood.
  • Formulations suitable for rectal administration are preferably presented as unit-dose suppositories. These may be prepared by admixing a compound of formula (I) with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
  • Formulations suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, transdermal patch, aerosol, or oil.
  • Carriers which may be used include vaseline, lanolin, polyethylene glycols, alcohols, and combinations of two or more thereof.
  • formulations may include other agents conventional in the art having regard to the type of formulation in question.
  • Synaptosomes for use in obtaining in vitro uptake data were prepared from hypothalamus or striatum by gently homogenizing the tissue in a 0.3 M sucrose/25 mM Tris pH 7.4 buffer containing iproniazid phosphate to inhibit monoamine oxidase. The homogenate was centrifuged at 1100 ⁇ g at 4° C. for 10 min and the supernatant was used for uptake studies. The supernatant (1 mg tissue protein) was incubated with Km concentrations of [ 3 H]-noradrenaline, [ 3 H]-dopamine or [ 3 H]-serotonin at 37° C.
  • Wellbutrin® was equipotent for inhibiting DA and noradrenaline uptake with IC 50 values of 1.9 and 2.2 ⁇ M, and did not inhibit serotonin uptake at 30 ⁇ M.
  • Imipramine (a non-specific tricyclic antidepressant) inhibited NA uptake and serotonin uptake with IC 50 values of 0.072 and 0.24 ⁇ M, respectively.
  • the compound of formula (I) was approximately twice as potent as Wellbutrin® as an NA inhibitor but, unlike the latter, was approximately 10-fold less potent as an inhibitor of dopamine uptake.
  • These data are consistent with the observed noradrenergic actions of Wellbutrin® and the racemic morpholinol metabolite of bupropion, (+/ ⁇ )-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride, (306U73) in vivo, at their respective anti-TBZ doses (Cooper, B. R., et al, Neuropsychopharmacology, 11: p. 133-41,1994). Behavioral and electrophysiological data suggest that the effects of Wellbutrin® are mediated by a noradrenergic mechanism (ibid).
  • Tetrabenazine (TBZ)-induced behavioural depression was used as an in vivo measure of antidepressant activity.
  • the test has been validated with a wide range of antidepressants, known to act through noradrenergic mechanisms (Cooper B. R. et al, “Animal models used in the prediction of antidepressant effects in man”, J. Clin. Psychiatry 44: 63-66, 1983).
  • the test was also used to identify Wellbutrin® as an anti-depressant. Briefly, animals were injected with the candidate agent (p.o. or i.p.) 30 minutes before receiving an i.p.
  • Results from the tetrabenazine-induced behavioural depression model are as follows. Assessed in vivo at 25 mg/kg (ip) the compound of formula (I), the racemate, Wellbutrin® and, for comparison, amitryptyline all abolished the tetrabenazine-induced behavioural depression. In contrast, the ( ⁇ ) enantiomer showed only modest activity (FIG. 1).
  • Wellbutrin® By extension, other activities of Wellbutrin® could be attributed to the compound of formula (I).
  • a noradrenergic mechanism is common to agents used to treat ADHD (e.g. methylphenidate and amphetamine). While the molecular mechanism for Wellbutrin's effects on smoking cessation is less well understood, a catecholaminergic pathway is thought to participate in the behavioural reinforcing properties of nicotine.
  • Wellbutrin® (and, by extension, the compound of formula (I)) by augmenting NA release into brain synapses, could mimic some of the actions of nicotine and, thus, decrease the signs associated with nicotine withdrawal. Additionally, amphetamines have been used to treat obesity.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Psychiatry (AREA)
  • Epidemiology (AREA)
  • Addiction (AREA)
  • Pain & Pain Management (AREA)
  • Psychology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Diabetes (AREA)
  • Child & Adolescent Psychology (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Disclosed is the compound (+)-(2S,3S)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol and pharmaceutically acceptable salts and solvates thereof, pharmaceutical compositions comprising them; also disclosed is a method of treating depression, attention deficit hyperactivity disorder (ADHD), obesity, migraine, pain, sexual dysfunction, Parkinson's disease, Alzheimer's disease, or addiction to cocaine or nicotine-containing (especially tobacco) products using such compound, salts, solvates or compositions.

Description

  • This invention relates to an optically pure morpholinol, pharmaceutical formulations containing it and processes for their preparation and use. [0001]
  • BACKGROUND OF THE INVENTION
  • Bupropion hydrochloride, (±)-1-(3-chlorophenyl)-2-[(1,1-dimethylethyl)-amino]-1-propanone hydrochloride, is the active ingredient of Wellbutrin® which is marketed in the United States for the treatment of depression. It is also the active ingredient of Zyban® which is marketed in the United States as an aid to smoking cessation. Bupropion is a relatively weak inhibitor of the neuronal uptake of noradrenaline (NA), serotonin and dopamine (DA), and does not inhibit monoamine oxidase. While the mechanism of action of bupropion, as with other antidepressants, is unknown, it is presumed that this action is mediated by noradrenergic and/or dopaminergic mechanisms. Available evidence suggests that Wellbutrin® is a selective inhibitor of noradrenaline (NA) at doses that are predictive of antidepressant activity in animal models. See Ascher, J. A., et al., Bupropion: A Review of its Mechanism of Antidepressant Activity. [0002] Journal of Clinical Psychiatry, 56: p. 395-401,1995.
    Figure US20020019396A1-20020214-C00001
  • Bupropion is extensively metabolized in man as well as laboratory animals. Urinary and plasma metabolites include biotransformation products formed via hydroxylation of the tert-butyl group and/or reduction of the carbonyl group of bupropion. Four basic metabolites have been identified. They are the erythro- and threo-amino alcohols of bupropion, the erythro-amino diol of bupropion, and a morpholinol metabolite. These metabolites of bupropion are pharmacologically active, but their potency and toxicity relative to bupropion have not been fully characterized. Because the plasma concentrations of the metabolites are higher than those of bupropion, they may be of clinical importance. [0003]
  • The morpholinol metabolite (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride is believed to be formed from hydroxylation of the tert-butyl group of bupropion. [0004]
    Figure US20020019396A1-20020214-C00002
  • SUMMARY OF THE INVENTION
  • It has now been discovered that despite the (−) form of the morpholinol metabolite predominating in human plasma samples, it is the (+) enantiomer, (+)-(2S, 3S)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol in which the activity resides. [0005]
  • Thus the present invention provides, in one aspect, a compound of formula (I), (+)-(2S, 3S)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol or pharmaceutically acceptable salts and solvates thereof. [0006]
    Figure US20020019396A1-20020214-C00003
  • Another aspect of the invention is pharmaceutical compositions comprising a compound of formula (I) or pharmaceutically acceptable salts and solvates thereof together with one or more pharmaceutically acceptable carriers, diluents or excipients. [0007]
  • A further aspect of the present invention is the use of a compound of formula (I) or pharmaceutically acceptable salts and solvates thereof in therapy. [0008]
  • Yet another aspect of the invention provides methods of treating depression, attention deficit hyperactivity disorder (ADHD), obesity, migraine, pain, sexual dysfunction, Parkinson's disease, Alzheimer's disease, or addiction to cocaine or tobacco products in a human or animal subject comprising the administration to said subject of an effective amount of a compound of formula (I) or pharmaceutically acceptable salts and solvates thereof or pharmaceutical compositions thereof. [0009]
  • Yet another aspect of the present invention is the use of the compound of formula (I) or pharmaceutically acceptable salts and solvates thereof or pharmaceutical compositions thereof in the preparation of a medicament for the treatment of depression, attention deficit hyperactivity disorder (ADHD), obesity, migraine, pain, sexual dysfunction, Parkinson's disease, Alzheimer's disease, addiction to cocaine or tobacco products. [0010]
  • DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Effect of Compounds at 25 mg/kg (ip) on TBZ-Induced Depression [0011]
  • FIG. 2. Dose Response of Compound of Formula I Against TBZ-Induced Depression (Compounds administered 30 minutes prior to TBZ, Male, CD-1 Mice, i.p., n=6) [0012]
  • FIG. 3. Dose Response of Compound of Formula II Against TBZ-Induced Depression (Compounds administered 30 minutes prior to TBZ, Male, CD-1 Mice, i.p., n=6)[0013]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The compound of formula (I) or pharmaceutically acceptable salts and solvates thereof may be prepared by first synthesizing the racemate of the morpholinol metabolite of bupropion and subsequently separating the (+) and (−) enantiomers of the racemate via HPLC. [0014]
  • The racemate of the morpholinol metabolite of bupropion hydrochloride ((+/+)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride) may be synthesized by the following process. To 3′-chloropropiophenone (10.0 g, 0.06 mol) in dioxane (50 mL) was added a solution of dioxane dibromide (14.9 g, 0.06 mol) in dioxane (50 mL). The reaction mixture was stirred for 2 h at ambient temperature and poured into a mixture of ice and water (500 mL). The mixture was extracted several times with methylene chloride. The combined extracts were dried (Na[0015] 2SO4) and concentrated in vacuo to give 14.8 g (85%) of 2-bromo-3′-chloropropiophenone as a pale yellow oil. This was used without further purification. NMR (300Mhz, CDCl3); δ7.99 (m, 1H)), 7.90 (d, 1H)), 7.57 (d, 1H)), 7.44 (t, 1H)), 5.22 (q, 1H)), 1.91 (t, 3H).
  • To a solution of 2-bromo-3′-chloropropiophenone (19.3 g, 0.08 mol) in MeOH (100 mL) was added dropwise a solution of 2-amino-2-methyl-1-propanol (27.8 g, 0.31 mol) in methanol (200 mL) at ambient temperature. The mixture was stirred for 18 h and concentrated in vacuo. The residue was partitioned between water and diethyl ether. The combined organic phase was extracted with 10% aqueous hydrogen chloride. The combined aqueous acid extracts were chilled in an ice bath and made basic with 40% aqueous sodium hydroxide. The mixture was extracted with diethyl ether, the combined diethyl ether extracts were washed with water and saturated sodium chloride solution, dried (K[0016] 2CO3) and concentrated in vacuo to give 15.0 g (75%) of (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol as an off-white solid.
  • (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol may be converted to (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride by the following process. A 6.0 g sample was dissolved in diethyl ether, chilled in an ice bath and ethereal hydrogen chloride added until the mixture was acidic. The resulting solid was filtered and recrystallized from ethanol/diethyl ether/ethereal hydrogen chloride mixtures to give 4.93 g of (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride as a white solid: m.p. 202-203° C. NMR (80 Mhz, DMSO-d[0017] 6); δ10.9 (br, 1H)), 8.85 (br, 1H)), 7.60-7.41 (m, 5H), 4.04 (d, 1H)), 3.50 (d, 1H)), 3.37 (br s, 1H)), 1.58 (s, 3H), 1.34 (s, 3H), 1.03 (d, 3H). Anal. Calcd for C13H19Cl2NO2:C, 53.43; H, 6.55; N, 4.79. Found: C, 53.54; H, 6.58; N, 4.75.
  • (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride may be converted back to its free base by the following process. A 3.0 g sample of (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride was dissolved in water (100 mL) and diethyl ether was added (200 mL). The mixture was chilled in an ice bath and the pH was adjusted to >10 with 1.0 N aqueous sodium hydroxide. After stirring for 30 min., the phases were separated and the aqueous phase was extracted with diethyl ether. The combined diethyl ether extracts were dried (Na[0018] 2SO4) and concentrated in vacuo to give 2.6 g of (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol as a white solid. This was used without further purification for the chiral chromatography described below.
  • The (+) and (−) enantiomers of (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol may be separated by the following process. (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol (2.54 gms.) was dissolved in 250 ml of 2:8 Isopropyl alcohol: Hexane (both HPLC grade). A Daicel Chiralcel OD column (2×25 cm.) was equilibrated for one hour at 8 ml./min. in the elution solvent, 1:9:0.2 Isopropanol:Hexane:Diethylamine. The solution of the (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol was injected in 1 ml. aliquots by an automated Waters Prep LC 2000, using a Waters 510 EF pump for injections. Each run was 15 minutes in length, using the conditions listed before. The separated optical isomers were collected by fraction collector (Waters) at a 2% above baseline threshold, based on 2 absorbance units full scale at 240 nm (Waters 490E UV detector). Each optical isomer solution was evaporated on a rotary evaporator at 40 degrees Centigrade and aspirator vacuum. After drying for 6 hours under high vacuum at room temperature, [0019] optical isomer 1 weighed 1.25 gm. and optical isomer 2 weighed 1.26 gm.
  • The enantiomeric purity of each isomer was assayed by analytical chiral HPLC on a Waters 860HPLC with 996 Photodiode Array detector, using a Daicel Chiralcel OD-H column (4.6×250 mm.) eluted with 1:9:0.2 Isopropyl alcohol: Hexane Diethylamine at 1 ml/min. [0020] Optical isomer 1 was 100% pure (R.T. 6.117 min.). Optical isomer 2 was 99.19% pure (R.T. 6.800 min.), containing 0.81% optical isomer 1 (R.T. 6.133 min.).
  • Hydrochloride salts of the separated enantiomers were obtained by the following processes. 1.25 g (0.005 mol) of optical isomer 1 (retention time 6.117 min) ((−)-(2R,3R)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol) was dissolved in diethyl ether. The solution was filtered and the filtrate was chilled in an ice-bath adding ethereal hydrogen chloride until the solution was acidic. After standing at ambient temperature for 24 h, the resulting solid was filtered, washed with diethyl ether and dried in a vacuum oven at 60° C. for 18 h to give 1.32 g (90%) of (−)-(2R,3R)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride as a white solid: mp 208-209° C. NMR (300Mhz, DMSO-d[0021] 6); δ9.72 (br, 1H)), 8.76 (br, 1H), 7.54-7.41 (m, 1H)), 3.98 (d, 1H)), 3.52 (d, 1H)), 3.37 (br s, 1H)), 1.53 (s, 1H)), 1.29 (s, 1H)), 0.97 (d, 1H)). Anal. Calcd for C13H19Cl2NO2:C, 53.43; H, 6.55; N, 4.79. Found: C, 53.35; H, 6.57; N, 4.71.
  • [α]D 20° C.=−33.2° (0.67, 95% EtOH)
  • 1.26 g (0.005 mol) of optical isomer 2 (retention time 6.800 min) (+)-(2S,3S)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol) was dissolved in diethyl ether. The solution was filtered and the filtrate was chilled in an ice-bath adding ethereal hydrogen chloride until the solution was acidic. After standing at ambient temperature for 24 h, the resulting solid was filtered, washed with diethyl ether and dried in a vacuum oven at 60° C. for 18 h to give 1.36 g (93%) of (+)-(2S,3S)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride as a white solid: mp 208-209° C. NMR (300Mhz, DMSO-d[0022] 6); δ9.87 (br, 1H)), 8.76 (br, 1H)), 7.54-7.41 (m, 1H)), 3.99 (d, 1H)), 3.51 (d, 1H)), 3.37 (br s, 1H)), 1.54 (s, 1H)), 1.30 (s, 1H)), 0.98 (d, 1H)). Anal. Calcd for C13H19Cl2NO2:C, 53.43; H, 6.55; N, 4.79. Found: C, 53.51; H, 6.58; N, 4.73.
  • [α]D 20° C.=+31.9° (0.64, 95% EtOH)
  • The absolute configuration of (+)-(2S,3S)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol was determined by the following x-ray crystallographic method. Crystal Data: C[0023] 13H18Cl2NO2, M=291, Orthorhombic, space group P212121, a=8.7348 (6), b=14.9824 (10), c=23.1605 (15) Å3, V=3031 (4) Å3, Z=8, Dc=1.276 Mgm−3, F(000)=1226.95. Of 12224 reflections measured. 3764 were unique and 2318 which had I>3.0σ(I) were used in subsequent calculations. Data was collected on a Siemens SMART diffractometer using omega scans and monochromated MoKα radiation (λ=0.71073 Å). The positions of all non-hydrogen atoms were determined by direct methods and refined anisotropically. The hydrogen positions were all located in difference syntheses and included in subsequent refinement cycles using a riding model and an idealized bond length of 0.96 Å. The absolute configuration was determined by refinement of the Rogers' parameter and confirmed by an analysis of the 185 best Bijvoet intensity differences which indicated a probability of 0.006 that the model was in error. Least squares refinement minimized Σw(ΔF)2 with weights based on counter statistics. The final agreement factors were Rf=0.064 (0.108 for all data), Rw=0.068 (0.081 for all data), and GoF=1.93. References included E. J. Gabe, Y. Le Page, J. -P. Charland, F. L. Lee and P. S. White, Journal of Applied Crystallography, 22, 384-387 (1989) and D. Rogers, Acta Crystallographica, A37, 734-741, 1981.
  • The amount of compound of formula (1) required to achieve the desired therapeutic effect will, of course depend on a number of factors, for example, the mode of administration, the recipient and the condition being treated. In general, the daily dose will be in the range of 0.02 to 5.0 mg/kg. More particular ranges include 0.02 to 2.5 mg/kg, 0.02 to 1.0 mg/kg, 0.02 to 0.25 mg/kg, 0.02 to 0.15 mg/kg and 0.02 to 0.07 mg/kg. [0024]
  • The compound of formula (I) may be employed in the treatment of depression, attention deficit hyperactivity disorder (ADHD), obesity, migraine, pain, sexual dysfunction, Parkinson's disease, Alzheimer's disease, addiction to cocaine or tobacco products as the compound per se, but is preferably presented with one or more pharmaceutically acceptable carriers, diluents or excipients in the form of a pharmaceutical formulation. The carriers, diluents and exipients must, of course, be acceptable in the sense of being compatible with the other ingredients of the formulation and must not be deleterious to the recipient. The carrier may be a solid or a liquid, or both, and is preferably formulated with the agent as a unit-dose formulation, for example, a tablet. [0025]
  • The formulations include those suitable for oral, rectal, topical, buccal (e.g. sub-lingual) and parenteral (e.g. subcutaneous, intramuscular, intradermal or intravenous) administration. [0026]
  • Formulations suitable for buccal (sub-lingual) administration include lozenges comprising a compound of formula (I) in a flavoured base, usually sucrose and acacia or tragacanth, and pastilles comprising the agent in an inert base such as gelatin and glycerin or sucrose and acacia. [0027]
  • Formulations of the present invention suitable for parenteral administration conveniently comprise sterile aqueous preparations of a compound of formula (I), preferably isotonic with the blood of the intended recipient. These preparations are preferably administered intravenously, although administration may also be effected by means of subcutaneous, intramuscular, or intradermal injection. Such preparations may conveniently be prepared by admixing the agent with water and rendering the resulting solution sterile and isotonic with the blood. [0028]
  • Formulations suitable for rectal administration are preferably presented as unit-dose suppositories. These may be prepared by admixing a compound of formula (I) with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture. [0029]
  • Formulations suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, transdermal patch, aerosol, or oil. Carriers which may be used include vaseline, lanolin, polyethylene glycols, alcohols, and combinations of two or more thereof. [0030]
  • It should be understood that in addition to the ingredients particularly mentioned above, the formulations may include other agents conventional in the art having regard to the type of formulation in question. [0031]
  • Biological activity of the compound of formula (I) was demonstrated by in vitro uptake models and the tetrabenazine-induced behavioural depression model. The racemic morpholinol metabolite, (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol, is referred to herein as “Racemate”. The (−) form of the morpholinol metabolite is (−)-(2R, 3R)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol or pharmaceutically acceptable salts and solvates thereof and is referred to herein as a compound of formula (II): [0032]
    Figure US20020019396A1-20020214-C00004
  • In vitro Synaptosomal Uptake Experiments. [0033]
  • In vitro uptake was determined, as reported previously, using synaptosomes prepared from rat caudoputamen (for dopamine uptake) and hypothalamus (for NA and serotonin uptake) using [[0034] 3H]-dopamine, [3H-]-NA and [3H-]-serotonin as transport substrates, respectively. See Eckhardt, S. B., R. A. Maxwell, and R. M. Ferris, A Structure-Activity Study of the Transport Sites for the Hypothalamic and Striatal Catecholamine Uptake Systems. Similarities and differences. Molecular Pharmacology, 21: p.374-9,1982.
  • Synaptosomes for use in obtaining in vitro uptake data were prepared from hypothalamus or striatum by gently homogenizing the tissue in a 0.3 M sucrose/25 mM Tris pH 7.4 buffer containing iproniazid phosphate to inhibit monoamine oxidase. The homogenate was centrifuged at 1100× g at 4° C. for 10 min and the supernatant was used for uptake studies. The supernatant (1 mg tissue protein) was incubated with Km concentrations of [[0035] 3H]-noradrenaline, [3H]-dopamine or [3H]-serotonin at 37° C. for 5 minutes in Modified Krebs-Henseleit buffer (118 mM NaCl, 5 mM KCl, 25 mM NaHCO3, 1.2 mM NaH2PO4, 1.2 MM MgSO4, 11 mM Dextrose, 2.5 MM CaCl2) in the absence and presence of drug. Under these conditions uptake was linear with respect to both for substrate and tissue (with <5% total substrate transported). Non-specific uptake was defined as uptake at 0° C. [3H]-substrate, which had been transported into synaptosomes, was separated from free [3H]-substrate by filtration over GF/B filters and washing with cold Krebs-Henseleit buffer. The filters were counted for tritum in a liquid scintillation spectrometer.
  • The data for in vitro synaptosomal uptake are presented as Table 1. Among the 2 enantiomers of the morpholinol metabolite of bupropion, the (+) enantiomer, the compound of formula (1), inhibited noradrenaline (NA) uptake with an IC[0036] 50 of 2.2 μM. In contrast, the (−) enantiomer was ineffective at a concentration of 30 μM. On dopamine (DA) uptake, the compound of formula (I) had an IC50 of ˜10 μM while the (−) enantiomer was inactive at 30 μM. Neither compound inhibited serotonin uptake at 30 mM.
  • For comparison, Wellbutrin® was equipotent for inhibiting DA and noradrenaline uptake with IC[0037] 50 values of 1.9 and 2.2 μM, and did not inhibit serotonin uptake at 30 μM. Imipramine (a non-specific tricyclic antidepressant) inhibited NA uptake and serotonin uptake with IC50 values of 0.072 and 0.24 μM, respectively.
  • The compound of formula (I) was approximately twice as potent as Wellbutrin® as an NA inhibitor but, unlike the latter, was approximately 10-fold less potent as an inhibitor of dopamine uptake. These data are consistent with the observed noradrenergic actions of Wellbutrin® and the racemic morpholinol metabolite of bupropion, (+/−)-(2R*,3R*)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol hydrochloride, (306U73) in vivo, at their respective anti-TBZ doses (Cooper, B. R., et al, [0038] Neuropsychopharmacology, 11: p. 133-41,1994). Behavioral and electrophysiological data suggest that the effects of Wellbutrin® are mediated by a noradrenergic mechanism (ibid).
  • Tetrabenazine-induced Behavioural Depression Experiments. [0039]
  • Tetrabenazine (TBZ)-induced behavioural depression was used as an in vivo measure of antidepressant activity. The test has been validated with a wide range of antidepressants, known to act through noradrenergic mechanisms (Cooper B. R. et al, “Animal models used in the prediction of antidepressant effects in man”, J. Clin. Psychiatry 44: 63-66, 1983). Moreover, the test was also used to identify Wellbutrin® as an anti-depressant. Briefly, animals were injected with the candidate agent (p.o. or i.p.) 30 minutes before receiving an i.p. injection of tetrabenazine (35 mg/kg, as the HCl salt—prepared fresh for each use). Assessments were performed 30 minutes thereafter and included: locomotor activity (1-4 scale); ptosis (1-4 scale) and body temperature as described previously (Cooper, B. R., J. L. Howard, and F. E. Soroko, Animal models used in prediction of antidepressant effects in man ([0040] Journal of Clinical Psychiatry, 44: p. 63-6,1983). In all studies, the scientist performing the assessments was blind to the treatments. All parameters were weighted equally to give a “lumped” score (X) through the following algorithm:
  • X=(1+Ptosis score)/(Activity score*[Temp,treated/Temp,control]
  • Results from the tetrabenazine-induced behavioural depression model are as follows. Assessed in vivo at 25 mg/kg (ip) the compound of formula (I), the racemate, Wellbutrin® and, for comparison, amitryptyline all abolished the tetrabenazine-induced behavioural depression. In contrast, the (−) enantiomer showed only modest activity (FIG. 1). [0041]
  • In the TBZ model of behavioural depression, activity resided in the compound of formula (I). When analysed in a dose-effect study with TBZ, the activity showed a sharp increase in activity between 3 mg/kg and 6 mg/kg (ip) (FIG. 2). The compound of formula II, in comparison, did not possess dose-related activity and, at 50 mg/kg, appeared to worsen the animal's condition (FIG. 3). In FIGS. 2 and 3, AMIT (5) refers to amitryptiline dosed at 5 mg/kg and SHAM refers to a control group of animals that have recieved no medication at all. [0042]
  • Since the TBZ test has been predictive of anti-depressants acting through noradrenergic mechanisms and the compound of formula (I) is an inhibitor of noradrenaline uptake and Wellbutrin® is metabolised to this morpholinol in vivo, the data suggest that the anti-depressant activity of Wellbutrin® is likely to result from the effects of the compound of formula (I). (Welch, R. M., A. A. Lai, and D. H. Schroeder, Pharmacological significance of the species differences in bupropion metabolism. [0043] Xenobiotica, 17: p. 287-98,1987).
  • By extension, other activities of Wellbutrin® could be attributed to the compound of formula (I). In particular, a noradrenergic mechanism is common to agents used to treat ADHD (e.g. methylphenidate and amphetamine). While the molecular mechanism for Wellbutrin's effects on smoking cessation is less well understood, a catecholaminergic pathway is thought to participate in the behavioural reinforcing properties of nicotine. Wellbutrin® (and, by extension, the compound of formula (I)), by augmenting NA release into brain synapses, could mimic some of the actions of nicotine and, thus, decrease the signs associated with nicotine withdrawal. Additionally, amphetamines have been used to treat obesity. The addictive properties of amphetamine, however, preclude its use for most obese patients. Wellbutrin® causes weight loss and, like amphetamine, acts through a noradrenergic mechanism. (Zarrindast, M. R. and T. Hosseini-Nia, Anorectic and behavioural effects of bupropion. General Pharmacology, 19: p. 201-4,1988 and Harto-Truax, N., et al., Effects of Bupropion on Body Weight. [0044] Journal of Clinical Psychiatry, 44: p. 183-6,1983). However, unlike amphetamine, Wellbutrin® is not addictive. (Lamb, R. J. and R. R. Griffiths, Self-administration in Baboons and the Discriminative Stimulus Effects in Rats of Bupropion, Nomifensine, Diclofensine and Imipramine. Psychopharmacology, 102: p. 183-90,1990; Bergman, J., et al., Effects of Cocaine and Related Drugs in Nonhuman Primates. III. Self-administration by Squirrel Monkeys. Journal of Pharmacology & Experimental Therapeutics, 251: p. 150-5,1989 and Johanson, C. E.; and J. E. Barrett, The Discriminative Stimulus Effects of Cocaine in Pigeons. Journal of Pharmacology & Experimental Therapeutics, 267: p. 1-8,1993). By extension, the compound of formula (I) would also be expected to have efficacy in obesity and cocaine addiction.
  • Safety and Toxicity. [0045]
  • Additional dose-ranging studies were performed to determine the range of safe does for the isomers and the racemate. Animals were observed for the presence of serious adverse events (e.g. seizures and deaths) following administration of the compounds of formula I, formula II or the racemate by the oral and intraperitoneal (i.p.) routes. The data are presented as Table II. [0046]
  • Administered orally, at 100 mg/kg p.o., seizures were observed with the compound of formula II and the racemate but not with the compound of formula I. Seizures were observed in all of the animals with all 3 compounds when dosed at 300 mg/kg. Additionally, at the 300 mg/kg oral dose resulted in 100 and 80% lethality for the compound of formula II and the racemate while no deaths were observed with the compound of formula I. [0047]
  • Administered i.p., all of the compounds produced seizures at 100 mg/kg. No deaths were observed with the compound of formula I whereas the compound of formula II and the racemate resulted in lethality of 100% and 20%, respectively. At the 300 mg/kg oral dose all of the lethality was observed for all of the compounds. [0048]
    TABLE 1
    Effects on Uptake In Vitro
    Compound IC50 (mM) SEM
    [3H]-Dopamine Uptake
    Bupropion 1.9 0.15
    Formula (I) 9.3 0.41
    Formula (II) >100
    [3H]-Noradrenaline Uptake
    Bupropion 2.2 0.7
    Formula (I) 1.1 0.07
    Formula (II) >30
    Imipramine 0.072 0.020
    [3H]-Serotonin Uptake
    Bupropion >30
    Formula (I) >30
    Formula (II) >100
    Imipramine 0.24 0.03
  • [0049]
    TABLE 2
    Adverse Events Associated with Compounds of Formula I, Formula II
    and the Racemate
    Time to Time to
    Dose Seizures Seizures Death
    Compound Route (mg/kg) (%) (min) % Died (min)
    Formula I i.p. 100 100 3.93 0 n/a
    Formula I p.o. 100  0 n/a 0 n/a
    Formula I i.p. 300 100 3.95 100 6
    Formula I p.o. 300 100 11.23 0 n/a
    Formula II i.p. 100  20 5 100 7
    Formula II p.o. 100 100 7.2 0 n/a
    Formula II i.p. 300 100 1.1 100 6
    Formula II p.o. 300 100 6.8 100 7
    Racemate i.p. 100 100 3 20 14 
    Racemate p.o. 100 100 9.2 0 n/a
    Racemate i.p. 300 100 3 100 3
    Racemate p.o. 300 100 6.8 80 7

Claims (1)

What is claimed is:
1. A method for inhibiting noradrenaline uptake, dopamine uptake or both comprising the administration of (+)-(2S,3S)-2-(3-chlorophenyl)-3,5,5-trimehtyl-2-morpholinol or pharmaceutically acceptable salts and solvates thereof.
US09/886,391 1998-01-21 2001-06-22 Pharmaceutically active morpholinol Expired - Fee Related US6391875B2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US09/886,391 US6391875B2 (en) 1998-01-21 2001-06-22 Pharmaceutically active morpholinol
US10/147,588 US6855820B2 (en) 1999-01-20 2002-05-17 Pharmaceutically active morpholinol
US10/150,339 US7098206B2 (en) 1998-01-21 2002-05-17 Pharmaceutically active morpholinol
US10/150,287 US6734213B2 (en) 1999-01-20 2002-05-17 Pharmaceutically active morpholinol
US10/150,341 US6998400B2 (en) 1998-01-22 2002-05-17 Pharmaceutically active morpholinol
US10/944,814 US20050096318A1 (en) 1999-01-20 2004-09-21 Pharmaceutically active morpholinol
US11/407,192 US20060189612A1 (en) 1998-01-21 2006-04-20 Pharmaceutically active morpholinol

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
GBGB9801230.5A GB9801230D0 (en) 1998-01-21 1998-01-21 Pharmaceutically active morpholinol
GB9801230 1998-01-21
GB9801230.5 1998-01-21
US7218098P 1998-01-22 1998-01-22
US09/233,531 US6274579B1 (en) 1998-01-21 1999-01-20 Pharmaceutically active morpholinol
US09/886,391 US6391875B2 (en) 1998-01-21 2001-06-22 Pharmaceutically active morpholinol

Related Parent Applications (4)

Application Number Title Priority Date Filing Date
US09/233,531 Division US6274579B1 (en) 1998-01-21 1999-01-20 Pharmaceutically active morpholinol
US09/233,531 Continuation US6274579B1 (en) 1998-01-21 1999-01-20 Pharmaceutically active morpholinol
PCT/US2000/010120 Continuation-In-Part WO2000061498A2 (en) 1996-07-15 2000-04-13 System for electrochemically processing a workpiece
US09/849,505 Continuation-In-Part US7020537B2 (en) 1999-04-13 2001-05-04 Tuning electrodes used in a reactor for electrochemically processing a microelectronic workpiece

Related Child Applications (6)

Application Number Title Priority Date Filing Date
US10/147,588 Continuation US6855820B2 (en) 1999-01-20 2002-05-17 Pharmaceutically active morpholinol
US10/147,588 Continuation-In-Part US6855820B2 (en) 1999-01-20 2002-05-17 Pharmaceutically active morpholinol
US10/150,287 Continuation-In-Part US6734213B2 (en) 1999-01-20 2002-05-17 Pharmaceutically active morpholinol
US10/150,341 Continuation-In-Part US6998400B2 (en) 1998-01-22 2002-05-17 Pharmaceutically active morpholinol
US10/150,339 Continuation-In-Part US7098206B2 (en) 1998-01-21 2002-05-17 Pharmaceutically active morpholinol
US11/097,508 Continuation US20050183959A1 (en) 2000-04-13 2005-03-31 Tuning electrodes used in a reactor for electrochemically processing a microelectric workpiece

Publications (2)

Publication Number Publication Date
US20020019396A1 true US20020019396A1 (en) 2002-02-14
US6391875B2 US6391875B2 (en) 2002-05-21

Family

ID=26312986

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/233,531 Expired - Fee Related US6274579B1 (en) 1998-01-21 1999-01-20 Pharmaceutically active morpholinol
US09/886,391 Expired - Fee Related US6391875B2 (en) 1998-01-21 2001-06-22 Pharmaceutically active morpholinol

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/233,531 Expired - Fee Related US6274579B1 (en) 1998-01-21 1999-01-20 Pharmaceutically active morpholinol

Country Status (30)

Country Link
US (2) US6274579B1 (en)
EP (2) EP1829544A1 (en)
JP (1) JP2002501025A (en)
KR (1) KR100568063B1 (en)
CN (2) CN1255389C (en)
AP (1) AP1229A (en)
AT (1) ATE365042T1 (en)
AU (1) AU755536B2 (en)
BR (1) BR9907203A (en)
CA (1) CA2318268A1 (en)
CY (1) CY1106828T1 (en)
DE (1) DE69936335T2 (en)
DK (1) DK1047428T3 (en)
EA (1) EA002410B1 (en)
EE (1) EE04452B1 (en)
ES (1) ES2288012T3 (en)
HR (2) HRP20000494B1 (en)
HU (1) HUP0100900A3 (en)
ID (1) ID26334A (en)
IL (2) IL137346A0 (en)
IS (1) IS2494B (en)
NO (2) NO326878B1 (en)
NZ (3) NZ520349A (en)
PL (1) PL193622B1 (en)
PT (1) PT1047428E (en)
SG (1) SG115489A1 (en)
SK (1) SK10912000A3 (en)
TR (1) TR200002126T2 (en)
WO (1) WO1999037305A1 (en)
YU (1) YU67102A (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020093649A1 (en) * 2000-12-21 2002-07-18 Jack Brass Method of producing an ultra-violet or near ultra-violet light source for non-destructive inspection or testing
US6979104B2 (en) 2001-12-31 2005-12-27 R.J. Doran & Co. LTD LED inspection lamp
US7546903B2 (en) 2005-02-04 2009-06-16 Kone Corporation Elevator system having location devices and sensors
US9510405B2 (en) 2011-06-30 2016-11-29 Schneider Electric Industries Sas Dual power SMPS for a modular lighting system

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7098206B2 (en) * 1998-01-21 2006-08-29 Smithkline Beecham Corporation Pharmaceutically active morpholinol
US6998400B2 (en) 1998-01-22 2006-02-14 Smithkline Beecham Corporation Pharmaceutically active morpholinol
WO1999038504A1 (en) 1998-01-29 1999-08-05 Sepracor Inc. Pharmaceutical uses of optically pure (-)-bupropion
WO1999038503A1 (en) 1998-01-29 1999-08-05 Sepracor Inc. Pharmacological uses of optically pure (+)-bupropion
US6855820B2 (en) 1999-01-20 2005-02-15 Smithkline Beecham Corporation Pharmaceutically active morpholinol
US6734213B2 (en) 1999-01-20 2004-05-11 Smithkline Beecham Corporation Pharmaceutically active morpholinol
US6342496B1 (en) * 1999-03-01 2002-01-29 Sepracor Inc. Bupropion metabolites and methods of use
US6337328B1 (en) * 1999-03-01 2002-01-08 Sepracor, Inc. Bupropion metabolites and methods of use
AU7750100A (en) * 1999-10-13 2001-04-23 Glaxo Group Limited Method for the treatment of obesity
GB0003232D0 (en) * 2000-02-11 2000-04-05 Smithkline Beecham Plc Novel composition
US20050215552A1 (en) * 2002-05-17 2005-09-29 Gadde Kishore M Method for treating obesity
US7109198B2 (en) 2002-05-17 2006-09-19 Duke University Method for treating obesity
GB0219687D0 (en) * 2002-08-23 2002-10-02 Lilly Co Eli Benzyl morpholine derivatives
ATE413882T1 (en) * 2002-08-23 2008-11-15 Lilly Co Eli 2-(PHENYLTHIOMETHYL)-MORPHOLINE DERIVATIVES FOR USE AS SELECTIVE NOREPINEPHRINE REUPPOSE INHIBITORS
CA2522708C (en) 2003-04-29 2013-05-28 Orexigen Therapeutics, Inc. Compositions for affecting weight loss
GB0325055D0 (en) * 2003-10-27 2003-12-03 Smithkline Beecham Corp New process
WO2005044809A1 (en) * 2003-10-27 2005-05-19 Smithkline Beecham Corporation Diastereomeric dynamic kinetic resolution process for preparing (+)-(2s, 3s)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol, salts, and solvates thereof
GB0326148D0 (en) * 2003-11-10 2003-12-17 Lilly Co Eli Morpholine derivatives
GB0327195D0 (en) * 2003-11-21 2003-12-24 Smithkline Beecham Corp Novel use
GB0327619D0 (en) * 2003-11-27 2003-12-31 Smithkline Beecham Corp New use
KR20060128995A (en) * 2004-01-13 2006-12-14 듀크 유니버시티 Compositions of an anticonvulsant and an antipsychotic drug for affecting weight loss
US7713959B2 (en) * 2004-01-13 2010-05-11 Duke University Compositions of an anticonvulsant and mirtazapine to prevent weight gain
WO2005110405A1 (en) * 2004-05-03 2005-11-24 Duke University Compositions for affecting weight loss
CN101370488B (en) 2005-11-22 2012-07-18 奥雷西根治疗公司 Compositions for increasing insulin sensitivity
CA2635261A1 (en) * 2006-01-03 2007-07-12 Algebra, Inc. Therapeutic amine-arylsulfonamide conjugate compounds
US8916195B2 (en) 2006-06-05 2014-12-23 Orexigen Therapeutics, Inc. Sustained release formulation of naltrexone
TWI504419B (en) 2006-11-09 2015-10-21 Orexigen Therapeutics Inc Layered pharmaceutical formulations
JP2010508997A (en) 2006-11-09 2010-03-25 オレキシジェン・セラピューティクス・インコーポレーテッド Methods for administering weight loss drugs
US20090075994A1 (en) * 2007-09-14 2009-03-19 Protia, Llc Deuterium-enriched radafaxine
WO2009158114A1 (en) 2008-05-30 2009-12-30 Orexigen Therapeutics, Inc. Methods for treating visceral fat conditions
EP2419400A1 (en) 2009-04-15 2012-02-22 Research Triangle Institute Monoamine reuptake inhibitors
RU2616496C2 (en) 2010-01-11 2017-04-17 Ориксиджен Терапьютикс, Инк. Methods of weight loss therapy in patients with dominant depression (versions)
JP2013526583A (en) 2010-05-21 2013-06-24 リサーチ・トライアングル・インスティチュート Phenylmorpholine and its analogs
JP5952267B2 (en) 2010-05-21 2016-07-13 リサーチ・トライアングル・インスティチュート Hydroxybupropion analogs to treat drug addiction (government-sponsored research or development)
CN102670617B (en) * 2012-04-13 2013-03-20 湖南大学 Application of ibuprofen 2-(2-arylmorpholine-4-yl) ethyl ester in preparation of antidepressant drugs
DK2858640T3 (en) 2012-06-06 2020-06-29 Nalpropion Pharmaceuticals Llc Composition for use in a method of treating obesity and obesity in patients with high cardiovascular risk
CN105949142B (en) * 2016-05-21 2018-03-27 南华大学 Single chiral compound with antidepressant activity and its preparation method and application
RU2763728C1 (en) * 2021-06-17 2021-12-30 Федеральное государственное автономное образовательное учреждение высшего образования "Пермский государственный национальный исследовательский университет" (ПГНИУ) Application of 3-(2-(4-nitrophenyl)-2-oxoethylidene)morpholin-2-one as antimicrobial agent

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US170430A (en) * 1875-11-30 Improvement in furnaces for heating steel in tempering
GB8417170D0 (en) 1984-07-05 1984-08-08 Wellcome Found Heterocyclic pharmaceutical compounds
GB9108629D0 (en) 1991-04-23 1991-06-12 Wellcome Found Heterocyclic pharmaceutical compounds,preparation and use
US6337328B1 (en) 1999-03-01 2002-01-08 Sepracor, Inc. Bupropion metabolites and methods of use

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020093649A1 (en) * 2000-12-21 2002-07-18 Jack Brass Method of producing an ultra-violet or near ultra-violet light source for non-destructive inspection or testing
US6979104B2 (en) 2001-12-31 2005-12-27 R.J. Doran & Co. LTD LED inspection lamp
US7546903B2 (en) 2005-02-04 2009-06-16 Kone Corporation Elevator system having location devices and sensors
US9510405B2 (en) 2011-06-30 2016-11-29 Schneider Electric Industries Sas Dual power SMPS for a modular lighting system

Also Published As

Publication number Publication date
NZ529316A (en) 2004-05-28
KR100568063B1 (en) 2006-04-07
DE69936335T2 (en) 2008-02-28
EA002410B1 (en) 2002-04-25
IS5568A (en) 2000-07-20
KR20010034285A (en) 2001-04-25
PT1047428E (en) 2007-09-07
HUP0100900A3 (en) 2002-08-28
EP1047428B1 (en) 2007-06-20
EP1047428A1 (en) 2000-11-02
YU67102A (en) 2004-12-31
AU2328099A (en) 1999-08-09
BR9907203A (en) 2000-10-17
CN1255389C (en) 2006-05-10
EP1047428A4 (en) 2001-12-19
DK1047428T3 (en) 2007-10-08
NO20003721D0 (en) 2000-07-20
TR200002126T2 (en) 2000-12-21
CY1106828T1 (en) 2012-05-23
EP1829544A1 (en) 2007-09-05
NZ520349A (en) 2004-02-27
ES2288012T3 (en) 2007-12-16
HRP20000494B1 (en) 2009-02-28
AU755536B2 (en) 2002-12-12
HRP20000494A2 (en) 2000-12-31
EE04452B1 (en) 2005-04-15
US6391875B2 (en) 2002-05-21
SK10912000A3 (en) 2001-04-09
EA200000691A1 (en) 2001-04-23
DE69936335D1 (en) 2007-08-02
HRP20051024A2 (en) 2006-04-30
IL161942A0 (en) 2005-11-20
WO1999037305A1 (en) 1999-07-29
AP1229A (en) 2003-12-04
NO20003721L (en) 2000-09-19
NO20083529L (en) 2000-09-19
NZ505809A (en) 2002-09-27
ID26334A (en) 2000-12-14
NO326878B1 (en) 2009-03-09
EE200000438A (en) 2001-12-17
CA2318268A1 (en) 1999-07-29
IS2494B (en) 2009-02-15
SG115489A1 (en) 2005-10-28
AP2000001869A0 (en) 2000-09-30
PL193622B1 (en) 2007-02-28
IL137346A0 (en) 2001-07-24
JP2002501025A (en) 2002-01-15
HUP0100900A2 (en) 2002-05-29
US6274579B1 (en) 2001-08-14
ATE365042T1 (en) 2007-07-15
CN1528753A (en) 2004-09-15
CN1294513A (en) 2001-05-09
CN1203858C (en) 2005-06-01
PL342012A1 (en) 2001-05-07

Similar Documents

Publication Publication Date Title
US6391875B2 (en) Pharmaceutically active morpholinol
US20060189612A1 (en) Pharmaceutically active morpholinol
US6855820B2 (en) Pharmaceutically active morpholinol
US6734213B2 (en) Pharmaceutically active morpholinol
US6998400B2 (en) Pharmaceutically active morpholinol
EP2094674B1 (en) A salt of 3-benzyl-2-methyl-2,3,3a,4,5,6,7,7a-octahydrobenzo[d]isoxazol-4-one
US20230150936A1 (en) Analogs of n,n,n-trimethyl-4-phosphoryloxytryptamine
AU2002315922B2 (en) Pharmaceutically active morpholinol
MXPA00007127A (en) Pharmaceutically active morpholinol
KR20040023653A (en) Pharmaceutically Active Morpholinol
CZ20002690A3 (en) Pharmaceutically acceptable salts and solvates of (+)-(2S,3S)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinol and morpholinol pharmaceutical preparation and pharmaceutical preparation
CN109912514A (en) (2- heteroaryl aminocarbonyl phenyl) azepine ring derivatives and application thereof
US20070004724A1 (en) (+)-(2S,3s)-2-(3-chlorophenyl)-3,5,5-trimethyl-2-morpholinor for treating anxiety
NZ715951B2 (en) Therapeutic agents for use in the prophylaxis and/or treatment of hyperkinetic movement disorders

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 4

FPAY Fee payment

Year of fee payment: 8

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20140521