EP0977572A1 - Use of nk-1 receptor antagonists for treating eating disorders - Google Patents

Use of nk-1 receptor antagonists for treating eating disorders

Info

Publication number
EP0977572A1
EP0977572A1 EP98917425A EP98917425A EP0977572A1 EP 0977572 A1 EP0977572 A1 EP 0977572A1 EP 98917425 A EP98917425 A EP 98917425A EP 98917425 A EP98917425 A EP 98917425A EP 0977572 A1 EP0977572 A1 EP 0977572A1
Authority
EP
European Patent Office
Prior art keywords
phenyl
alkyl
group
hydrogen
substituted
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.)
Withdrawn
Application number
EP98917425A
Other languages
German (de)
French (fr)
Inventor
Franz F. Hefti
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.)
Organon Pharma UK Ltd
Original Assignee
Merck Sharp and Dohme 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
Priority claimed from GBGB9708289.5A external-priority patent/GB9708289D0/en
Priority claimed from GBGB9721265.8A external-priority patent/GB9721265D0/en
Application filed by Merck Sharp and Dohme Ltd filed Critical Merck Sharp and Dohme Ltd
Publication of EP0977572A1 publication Critical patent/EP0977572A1/en
Withdrawn legal-status Critical Current

Links

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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/4035Isoindoles, e.g. phthalimide
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/438The ring being spiro-condensed with carbocyclic or heterocyclic ring systems
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/451Non condensed piperidines, e.g. piperocaine having a carbocyclic group directly attached to the heterocyclic ring, e.g. glutethimide, meperidine, loperamide, phencyclidine, piminodine
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • 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
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • This invention relates to the treatment or prevention of eating disorders by the administration of a NK-1 receptor antagonist, optionally in combination with an anorectic agent.
  • Eating disorders commonly arise through an imbalance in a subject's appetite, or desire to eat. It is recognised that appetite is influenced by the interaction of central and peripheral systems, most likely acting through the effects of neuropeptides on the so-called feeding and saiety centres in the hypothalamus region of the brain. For instance, neuropeptides released by the gut in response to a meal may serve to modulate the intake of further food.
  • Alterations of appetite may lead to eating disorders including obesity, bulimia nervosa, and compulsive eating disorders.
  • Obesity is a chronic disease that is highly prevalent in modern society and is associated not only with a social stigma, but also with decreased life span and numerous medical problems, including adverse psychological development, reproductive disorders such as polycystic ovarian disease, dermatological disorders such as infections, varicose veins, Acanthosis nigricans, and eczema, exercise intolerance, diabetes mellitus, insulin resistance, hypertension, hypercholesterolemia, cholelithiasis, osteoarthritis, orthopedic injury, thromboembolic disease, cancer, and coronary heart disease. Rissanen et al, British Medical Journal, 301:835-837 (1990).
  • Bulimia nervosa is characterised by recurrent episodes of overeating, or binges, followed by severe dieting often associated with self- induced vomiting, abuse of laxatives or diuretics, or excessive exercise to avoid weight gain. Frequent vomiting or purging may result in electrolyte disturbances and erosion of dental enamel.
  • Complusive eating disorders may or may not be associated with other neurological disorders.
  • One well characterised compulsive eating disorder is Prader-Willi syndrome, a congential disorder characterised by infantile hypotonia, hypogonadism and facial dysmorphism, with subsequent development of hyperplagia and abnormalities of behaviour and intellect.
  • Treatment regimens for eating disorders typically include the use of anorectic agents, such as amphetamine derivatives.
  • anorectic agents such as amphetamine derivatives.
  • jo-Chloroamphetamine and other halogenated amphetamines promote serotonin (5-hydroxytryptamine; 5-HT) release from platelets and neurons.
  • a rapid release of serotonin is followed by a prolonged and selective depletion of serotonin in the brain.
  • the most widely used example of this class of compound is fenfluramine and its (S)-isomer, dexfenfluramine.
  • fenfluramine and dexfenfluramine are both useful in the treatment of bulimia nervosa and obesity, and fenfluramine has also produced promising results in the management of Prader-Willi syndrome.
  • Neurokinin 1 (NK-1; substance P) receptor antagonists are being developed for the treatment of a number of physiological disorders associated with an excess or imbalance of tachykinins, and in particular substance P. Examples of such conditions include disorders of the central nervous system such as anxiety, depression and psychosis (see, for instance, International (PCT) patent specification Nos. WO 95/16679, WO 95/18124 and WO 95/23798).
  • NK-1 receptor antagonists are effective in the treatment of eating disorders, as evidenced by their activity in vivo in a model of diet-induced obesity.
  • a combination of an anorectic agent with a NK-1 receptor antagonist may provide an enhanced anorectic effect. They may also provide for a rapid onset of action to combat eating disorders thereby enabling prescription on an "as-needed" basis.
  • a particularly preferred class of NK-1 receptor antagonists of use in the present invention are those which are able to cross the blood-brain barrier, otherwise known as CNS- or brain-penetrant compounds. The present invention accordingly provides the use of a NK-1 receptor antagonist for the manufacture of a medicament for the treatment or prevention of eating disorders.
  • the present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist.
  • a pharmaceutical composition for the treatment or prevention of eating disorders comprising a NK-1 receptor antagonist, together with at least one pharmaceutically acceptable carrier or excipient.
  • a NK-1 receptor antagonist for the manufacture of a medicament for the treatment or prevention of obesity.
  • the present invention also provides a method for the treatment or prevention of obesity, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist.
  • a NK-1 receptor antagonist for the manufacture of a medicament for the treatment or prevention of bulimia nervosa.
  • the present invention also provides a method for the treatment or prevention of bulimia nervosa, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist.
  • a NK-1 receptor antagonist for the manufacture of a medicament for the treatment or prevention of compulsive eating disorders.
  • the present invention also provides a method for the treatment or prevention of compulsive eating disorders, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist.
  • a NK-1 receptor antagonist for the manufacture of a medicament for reducing the total body fat mass in an obese mammal, especially a human.
  • the present invention also provides a method for reducing the total body fat mass in an obese mammal, especially a human, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist.
  • the present invention further provides the use of a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for the treatment or prevention of eating disorders.
  • the present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
  • a pharmaceutical composition comprising a NK-1 receptor antagonist and an anorectic agent, together with at least one pharmaceutically acceptable carrier or excipient.
  • the NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of eating disorders. Such combined preparations may be, for example, in the form of a twin pack.
  • a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
  • the use of a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for the treatment or prevention of obesity is also provides a method for the treatment or prevention of obesity, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
  • NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of obesity.
  • Such combined preparations may be, for example, in the form of a twin pack.
  • a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of obesity.
  • a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for the treatment or prevention of bulimia nervosa.
  • the present invention also provides a method for the treatment or prevention of bulimia nervosa, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
  • the NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of bulimia nervosa. Such combined preparations may be, for example, in the form of a twin pack.
  • a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of bulimia nervosa.
  • a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for the treatment or prevention of compulsive eating disorders.
  • the present invention also provides a method for the treatment or prevention of compulsive eating disorders, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
  • NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of compulsive eating disorders.
  • Such combined preparations may be, for example, in the form of a twin pack.
  • a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of compulsive eating disorders.
  • a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for reducing the total body fat mass in an obese mammal, especially a human.
  • the present invention also provides a method for reducing the total body fat mass in an obese mammal, especially a human, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
  • NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for reducing the total body fat mass in an obese mammal, especially a human.
  • Such combined preparations may be, for example, in the form of a twin pack.
  • a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in reducing the total body fat mass in an obese mammal, especially a human.
  • both the NK-1 receptor antagonist and the anorectic agent will be administered to a patient, within a reasonable period of time.
  • the compounds may be in the same pharmaceutically acceptable carrier and therefore administered simultaneously. They may be in separate pharmaceutical carriers such as conventional oral dosage forms which are taken simultaneously.
  • the term "combination" also refers to the case where the compounds are provided in separate dosage forms and are administered sequentially. Therefore, by way of example, the anorectic agent may be administered as a tablet and then, within a reasonable period of time, the NK-1 receptor antagonist may be administered either as an oral dosage form such as a tablet or a fast-dissolving oral dosage form.
  • a “fast dissolving oral formulation” is meant, an oral delivery form which when placed on the tongue of a patient, dissolves within about 10 seconds.
  • “reasonable period of time” is meant a time period that is not in excess of about 1 hour. That is, for example, if the anorectic agent is provided as a tablet, then within one hour, the NK-1 receptor antagonist should be administered, either in the same type of dosage form, or another dosage form which provides effective delivery of the medicament.
  • the compositions of the present invention are useful for the prevention or treatment of eating disorders.
  • eating disorders includes obesity, bulimia nervosa and compulsive eating disorders.
  • obesity refers to a condition whereby a mammal has a Body Mass Index (BMI), which is calculated as weight per height squared (kg/m 2 ), of at least 25.9. Conventionally, those persons with normal weight, have a BMI of 19.9 to less than 25.9.
  • BMI Body Mass Index
  • the obesity herein may be due to any cause, whether genetic or environmental.
  • disorders that may result in obesity or be the cause of obesity include overeating and bulimia, polycystic ovarian disease, craniopharyngioma, the Prader-Willi Syndrome, Frohlich's syndrome, Type II diabetes, GH-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g, children with acute lymphoblastic leukemia.
  • Treatment refers to reducing the BMI of the mammal to less than about 25.9, and maintaining that weight for at least 6 months.
  • the treatment suitably results in a reduction in food or calorie intake by the mammal.
  • Prevention refers to preventing obesity from occurring if the treatment is administered prior to the onset of the obese condition. Moreover, if treatment is commenced in already obese subjects, such treatment is expected to prevent, or to prevent the progression of, the medical sequelae of obesity, such as, e.g., arteriosclerosis, Type II diabetes, polycycstic ovarian disease, cardiovascular diseases, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, and cholelithiasis.
  • this invention relates to the inhibition and/or complete suppression of lipogenesis in obese mammals, i.e., the excessive accumulation of lipids in fat cells, which is one of the major features of human and animal obesity, as well as loss of total body weight.
  • the invention ameliorates the conditions that are a consequence of the disease, such as preventing or arresting the progression of polycystic ovarian disease so that the patient is no longer infertile, and increasing the insulin sensitivity and/or decreasing or eliminating the need or usage of insulin in a diabetic patient, e.g., one with adult-onset diabetes or Type II diabetes.
  • mammals include animals of economic importance such as bovine, ovine, and porcine animals, especially those that produce meat, as well as domestic animals, sports animals, zoo animals, and humans, the latter being preferred.
  • compositions of the present invention are especially useful for the treatment of or prevention of eating disorders where the use of an anorectic agent is generally prescribed.
  • an anorectic agent is generally prescribed.
  • Suitable anoretic agents of use in the combinations of the present invention include, but are not limited to, aminorex, amphechloral, amphetamine, benzphetamine, chlorphentermine, clobenzorex, cloforex, clominorex, clortermine, cyclexedrine, dexfenfluramine, dextroamphetamine, diethylpropion, diphemethoxidine, N- ethylamphetamine, fenbutrazate, fenfluramine, fenisorex, fenproporex, fludorex, fluminorex, furfurylmethylamphetamine, levamfetamine, levophacetoperane, mazindol, mefenorex, metamfepramone, methamphetamine, norpseudoephedrine, pentorex, phendimetrazine, phenmetrazine, phentermine, phenylpropan
  • anorectic agents include amphetamine and derivatives thereof such as amphetamine, benzphetamine, chlorphentermine, clobenzorex, cloforex, clotermine, dexfenfluramine, dextroamphetamine, diethylpropion, N-ethylamphetamine, fenfluramine, fenproporex, furfurylmethylamphetamine, levamfetamine, mefenorex, metamfepramone, methamphetamine, norpseudoephedrine, pentorex, phendimetrazine, phenmetrazine, phentermine, phenylpropanolamine, picilorex and sibutramine; and pharmaceutically acceptable salts thereof.
  • amphetamine and derivatives thereof such as amphetamine, benzphetamine, chlorphentermine, clobenzorex, cloforex, clotermine, dexfenfluramine, dex
  • a particularly suitable class of anorectic agent are the halogenated amphetamine derivatives, including chlorphentermine, cloforex, clortermine, dexfenfluramine, fenfluramine, picilorex and sibutramine; and pharmaceutically acceptble salts thereof;
  • halogenated amphetamine derivatives of use in the present invention include: fenfluramine and dexfenfluramine, and pharmaceutically acceptable salts thereof.
  • Another particularly preferred anorectic agent is phentermine.
  • NK-1 receptor antagonists of use in the present invention are described in published European Patent Specification Nos. 0 360 390, 0 394 989, 0 429 366, 0 443 132, 0 482 539, 0 512 901, 0 512 902, 0 514 273, 0 514 275, 0 517 589, 0 520 555, 0 522 808, 0 528 495, 0 532 456, 0 533 280, 0 536 817, 0 545 478, 0 577 394, 0 590 152, 0 599 538, 0 610 793, 0 634 402, 0 686 629, 0 693 489, 0 694 535, 0 699 655, 0 699 674, 0 707 006, 0 708 101, 0 714 891, 0 723 959, 0 733 632 and 0 776 893; and in International Patent Specification Nos
  • NK-1 receptor antagonists are those described in European Patent Specification No. 0 577 394, i.e. compounds of formula (I):
  • R 1 is selected from the group consisting of:
  • heterocycle wherein the heterocycle is selected from the group consisting of:
  • heterocylcle is unsubstituted or substituted with one or more substituent(s) selected from: (i) Ci-ealkyl, unsubstituted or substituted with halo, -CF3,
  • R 9 is as defined above;
  • R 2 and R 3 are independently selected from the group consisting of:
  • R 1 and R 2 may be joined together to form a heterocyclic ring selected from the group consisting of: (a) pyrrolidinyl, (b) piperidinyl,
  • R 2 and R 3 may be joined together to form a heterocyclic ring selected from the group consisting of: (a) pyrrolidinyl, (b) piperidinyl,
  • Ci-ealkyl (i) Ci-ealkyl, (ii) oxo, (iii) Ci-calkoxy,
  • X is selected from the group consisting of:
  • R 4 is selected from the group consisting of: (1)
  • alkyl is unsubstituted or substituted with one or more of the substituents selected from: (a) hydroxy,
  • R 5 is selected from the group consisting of: (1) phenyl, unsubstituted or substituted with one or more of Ru, R i2 and R 13 ; (2) Ci-salkyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
  • R G , R 7 and R 8 are independently selected from the group consisting of: (1) hydrogen; (2) Ci- ⁇ alkyl, unsubstituted or substituted with one or more of the substituents selected from:
  • Y is selected from the group consisting of: (1) a single bond,
  • R 15 and R i6 are independently selected from the group consisting of:
  • Ci-ealkyl unsubstituted or substituted with one or more of the substituents selected from: (i) hydroxy,
  • Z is selected from:
  • R is selected from the group consisting of:
  • Ci- ⁇ alkyl substituted with one or more of the substituents selected from:
  • heterocycle wherein the heterocycle is selected from the group consisting of:
  • Ci- ⁇ alkyl unsubstituted or substituted with halo, -CF 3 , -OCH3, or phenyl
  • R 2 and R 3 are independently selected from the group consisting of: (1) hydrogen;
  • X is - 0-
  • R 5 is phenyl, unsubstituted or substituted with halo;
  • R 6 , R 7 and R 8 are independently selected from the group consisting of: (1) hydrogen,
  • NK-1 receptor antagonists are those described in International (PCT) Patent Specification No. WO 95/18124, i.e. compounds of formula (II):
  • R i is hydrogen, halogen, Ci- ⁇ alkyl, d-ealkoxy, CF 3) NO2, CN, SR a , SOR a , SO 2 R a , CO 2 R a , CONR a R b , C 2 -6alkenyl, C 2 - 6 alkynyl or C 1 . 4 alkyl substituted by C ⁇ - 4 alkoxy, where R a and R b each independently represent hydrogen or Cj.. alkyl;
  • R 2 is hydrogen, halogen, Ci-ealkyl, Ci-ealkoxy substituted by Ci. 4 alkoxy or CF3;
  • R 3 is hydrogen, halogen or CF3
  • R 4 is hydrogen, halogen, Ci- ⁇ alkyl, Ci- ⁇ alkoxy, CF 3 , N0 2 , CN, SR a , SOR a , SO 2 R a , CO 2 R a , CONR a R b , or C ⁇ . alkyl substituted by Ci- alkoxy, where R a and R b each independently represent hydrogen or Ci- alkyl;
  • R 5 is hydrogen, halogen, Ci-ealkyl, Ci-ealkoxy substituted by C ⁇ - alkoxy or CF3;
  • R 6 is a 5-membered or 6-membered heterocyclic ring containing 2 or
  • R 7 is hydrogen, C ⁇ - alkyl, C3-7cycloalkyl or C3-7cycloalkylCi- 4 alkyl, or C 2 - alkyl substituted by C ⁇ -4alkoxy or hydroxyl;
  • R 8 is hydrogen, C ⁇ .
  • R 7 , R 8 and the nitrogen atom to which they are attached form a non-aromatic azabicyclic ring system of 6 to 12 ring atoms; or Z, R 7 and the nitrogen atom to which they are attached form a heteroaliphatic ring of 4 to 7 ring atoms which may optionally contain an oxygen ring atom;
  • R 9a and R 9b are each independently hydrogen or Ci- alkyl, or R 9a and R 9b are joined so, together with the carbon atoms to which they are attached, there is formed a C5-7 ring;
  • X is an alkylene chain of 1 to 4 carbon atoms optionally substituted by oxo
  • Y is a C ⁇ - 4 alkyl group optionally substituted by a hydroxyl group; with the proviso that if Y is C ⁇ - 4 alkyl, R 6 is susbstituted at least by a group of formula ZNR 7 R 8 as defined above.
  • Particularly preferred compounds of formula (II) are those of formula (Ila) and pharmaceutically acceptable salts thereof:
  • a 1 is fluorine or CF3
  • a 2 is fluorine or CF3
  • a 3 is fluorine or hydrogen; and X, Y and R 6 are as defined in relation to formula (II).
  • Particularly preferred compounds of formula (II) include: 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(5-(dimethylamino) methyl-l,2,3-triazol-4-yl)methyl-3-(S)-phenylmorpholine; 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(5-(dimethylamino) methyl-l,2,3-triazol-4-yl)methyl-3-(S)-(4-fluorophenyl)morpholine; and pharmaceutically acceptable salts thereof.
  • NK-1 receptor antagonists are those described in European Patent Specification No. WO 95/23798, i.e. compounds of formula (III):
  • R 2 and R 3 are independently selected from the group consisting of:
  • Ci-ealkyl unsubstituted or substituted with one or more of the substituents selected from: (a) hydroxy,
  • R 2 and R 3 may be joined together to form a heterocyclic ring selected from the group consisting of:
  • R 6 , R 7 and R 8 are independently selected from the group consisting of:
  • Ci- ⁇ alkyl unsubstituted or substituted with one or more of the substituents selected from: (a) hydroxy,
  • R U , R i2 and R i3 are independently selected from the definitions of R 6 , R 7 and R 8 , or -OX;
  • A is selected from the group consisting of:
  • Ci- ⁇ alkyl unsubstituted or substituted with one or more of the substituents selected from:
  • B is a heterocycle, wherein the heterocycle is selected from the group consisting of:
  • heterocycle may be substituted in addition to -X with one or more substituent(s) selected from:
  • Ci- ⁇ alkyl unsubstituted or substituted with halo, -CF 3 , -OCH3, or phenyl
  • p is 0 or 1;
  • X is selected from:
  • Y is selected from the group consisting of:
  • R 15 and R 1G are independently selected from the group consisting of: (a) Ci- ⁇ alkyl, unsubstituted or substituted with one or more of the substituents selected from:
  • Z is selected from:
  • R 2 and R 3 are independently selected from the group consisting of:
  • R 6 , R 7 and R 8 are independently selected from the group consisting of:
  • Ru, R i2 and R X3 are independently selected from the group consisting of: (1) fluoro, (2) chloro,
  • A is unsubstituted l-ealkyl
  • B is selected from the group consisting of:
  • p is 0 or 1;
  • X is selected from:
  • R 5 is selected from the group consisting of:
  • Y is -O-
  • Z is hydrogen or Ci-calkyl; and pharmaceutically acceptable salts thereof.
  • Particularly preferred compounds of formula (III) include:
  • NK-1 receptor antagonists are those described in European Patent Specification No. WO 96/05181, i.e. compounds of formula (IV):
  • X is a group of the formula ⁇ R 6 R 7 or a C- or N-linked imidazolyl ring;
  • Y is hydrogen or C 1 . 4 alkyl optionally substituted by a hydroxy group
  • R is hydrogen, halogen, d-ealkyl, d-ealkoxy, CF 3 , NO 2 , CN, SR a , SOR a , SO 2 R a , CO 2 R a , CONR a R b , C ⁇ alkenyl, C 2 -Galkynyl or d- alkyl substituted by d- alkoxy, wherein R a and R b each independently represent hydrogen or C ⁇ - alkyl;
  • R 2 is hydrogen, halogen, d- ⁇ alkyl, Ci-ealkoxy substituted by Ci- alkoxy or CF 3 ;
  • R 3 is hydrogen, halogen or CF3;
  • R 4 is hydrogen, halogen, d-ealkyl, C 1 .6alkoxy, hydroxy, CF 3 , NO2,
  • R 5 is hydrogen, halogen, Ci-Galkyl, d-6alkoxy substituted by C ⁇ _ 4 alkoxy or CF 3 ;
  • R G is hydrogen, d-Galkyl, C 3 -7cycloalkyl, C3-7cycloalkylC ⁇ - alkyl, phenyl, or C2-4alkyl substituted by Ci- 4 alkoxy or hydroxy;
  • R 7 is hydrogen, d-ealkyl, C 3 . 7 cycloalkyl, C 3 -7cycloalkylC ⁇ -4alkyl, phenyl, or C 2 . alkyl substituted by one or two substituents selected from Ci- 4 alkoxy, hydroxy or a 4, 5 or 6 membered heteroaliphatic ring containing one or two heteroatoms selected from N, O and S; or R 6 and R 7 , together with the nitrogen atom to which they are attached, form a saturated or partially saturated heterocyclic ring of 4 to 7 ring atoms, which ring may optionally contain in the ring one oxygen or sulphur atom or a group selected from NR 8 , S(O) or S(O)2 and which ring may be optionally substituted by one or two groups selected from hydroxyC 1 .
  • R 8 is hydrogen, d- 4 alkyl, hydroxy Ci- alkyl or d-4alkoxyd.4alkyl; and R 9a and R 9b are each independently hydrogen or C 1 . alkyl, or R 9a and
  • R 9b are joined so, together with the carbon atoms to which they are attached, there is formed a C5-7 ring; and pharmaceutically acceptable salts thereof.
  • Particularly preferred compounds of formula (IV) are those of formula (IVa) and pharmaceutically acceptable salts thereof:
  • a 1 is fluorine or CF3;
  • a 2 is fluorine or CF3;
  • a 3 is fluorine or hydrogen; and X and Y are as defined in relation to formula (I).
  • NK-1 receptor antagonists of use in the present invention is that described in European Patent Specification No.
  • Y is (CH 2 )n wherein n is an integer from 1 to 4, and wherein any one of the carbon-carbon single bonds in said (CH2)n may optionally be replaced by a carbon-carbon double bond, and wherein any one of the carbon atoms of said (CH2)n may optionally be substituted with R 4 , and wherein any one of the carbon atoms of said (CH2)n may optionally be substituted with R 7 ;
  • Z is (CH2)m wherein m is an integer from 0 to 6, and wherein any one of the carbon-carbon single bonds of (CH2) may optionally be replaced by a carbon-carbon double bond or a carbon-carbon triple bond, and any one of the carbon atoms of said (CH 2 ) m may optionally be substituted with R 8 ;
  • R i is hydrogen or Ci-salkyl optionally substituted with hydroxy, C ⁇ - 4 alkoxy or fluoro;
  • R 2 is a radical selected from hydrogen, d-e straight or branched alkyl, C3-7cycloalkyl wherein one of the CH 2 groups in said cycloalkyl may optionally be replaced by NH, oxygen or sulphur; aryl selected from phenyl and naphthyl; heteroaryl selected from indanyl, thienyl, furyl, pyridyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl and quinolyl; phenyl- C2-6alkyl, benzhydryl and benzyl, wherein each of said aryl and heteroaryl groups and the phenyl moieties of said benzyl, phenyl - C 2 -6 lkyl and benzhydr
  • R 4 and R 7 are each independently selected from hydroxy, halogen, halo, amino, oxo, cyano, methylene, hydroxymethyl, halomethyl, Ci-ealkylamino, di-d-ealkylamino, Ci-ealkoxy, C ⁇ -6alkyl-O-CO, Ci-ealkyl-O-CO-d-ealkyl, Ci-ealkyl-CO-O, Ci-ealkyl-CO-Ci-ealkyl-O-, Ci-ealkyl-CO-, Ci-Galkyl-CO-d-ealkyl, and the radicals set forth in the definition of R 2 ;
  • Ro is -NHCOR 9 , -NHCH2R 9 , SO2R 8 or one of the radicals set forth in any of the definitions of R 2 , R 4 and R 7 ;
  • R 9 is Ci-ealkyl, hydrogen, phenyl or phenylC ⁇ -6alkyl; with the proviso that (a) when m is 0, R 8 is absent, (b) when R 4 , R G , R 7 or R 8 is as defined in R 2 , it cannot form together with the carbon to which it is attached ,a ring with R 5 , and (c) when R 4 and R 7 are attached to the same carbon atom, then either each of R 4 and R 7 is independently selected from hydrogen, fluoro and Ci-ealkyl, or R 4 and R 7 , together with the carbon to which they are attached, for a C3-6 saturated carbocyclic ring that forms a spiro compound with the nitrogen-containing ring to which they are attached.
  • a particularly preferred compound of formula (V) is (2S,3S)-cis-3-(2- methoxybenzylamino)-2-phenylpiperidine; or a pharmaceutically acceptable salt thereof.
  • NK-1 receptor antagonists of use in the present invention is that described in International Patent Specification No. WO 93/21155, i.e. compounds of formula (VI):
  • radicals R are phenyl radicals optionally 2- or 3-substituted by a halogen atom or a methyl radical;
  • R i is optionally substituted phenyl, cyclohexadienyl, naphthyl, indenyl or optionally substituted heterocycle;
  • R 2 is H, halogen, OH, alkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, alkyloxy, alkylthio, acyloxy, carboxy, optionally substituted alkyloxycarbonyl, benzyloxycarbonyl, amino or acylamino;
  • R 3 is optionally 2-substituted phenyl;
  • R 4 is OH or fluorine when R 5 is H; or R 4 and R 5 are OH ; or R 4 and R 5 together form a bond.
  • a particularly preferred compound of formula (VI) is (3aS, 4S, 7aS)- 7,7-diphenyl-4-(2-methoxyphenyl)-2-[(2S)-(2-methoxyphenyl)propionyl] perhydroisoindol-4-ol; or a pharmaceutically acceptable salt thereof.
  • Another class of NK-1 receptor antagonists of use in the present invention is that described in European Patent Specification No. 0 591 040, i.e. compounds of formula (VII):
  • Ar represents an optionally substituted mono-, di- or tricyclic aromatic or heteroaromatic group
  • T represents a bond, a hydroxymethylene group, a C ⁇ - 4 alkoxymethylene group or a Ci-salkylene group
  • Ar' represents a phenyl group which is unsubstituted or substituted by one or more substituents selected from halogen, preferably chlorine or fluorine, trifluoromethyl, d- 4 alkoxy, C ⁇ - 4 alkyl where the said substituents may be the same or different; a thienyl group; a benzothienyl group; a naphthyl group; or an indolyl group;
  • R represents hydrogen, C ⁇ . alkyl, ⁇ -C ⁇ . alkoxyC ⁇ - alkyl, or ⁇ - C 2 - 4 alkanoyloxy C2-4alkyl;
  • Q represents hydrogen; or Q and R together form a 1,2-ethylene, 1,3-propylene or 1,4- butylene group;
  • Am + represents the radical
  • a particularly preferred compound of formula (VII) is (+) l-[2-[3- (3,4-dichlorophenyl)-l-[(3-isopropoxyphenyl)acetyl]-3-piperidinyl]ethyl]-4- phenyl-l-azabicyclo[2,2,2]octane; or a pharmaceutically acceptable salt, especially the chloride, thereof.
  • Another class of NK-1 receptor antagonists of use in the present invention is that described in European Patent Specification No. 0 532 456, i.e. compounds of formula (VIII):
  • R i represents an optionally substituted aralkyl, aryloxyalykl, heteroaralkyl, aroyl, heteroaroyl, cycloalkylcarbonyl, aralkanoyl, heteroarylalkanoyl, aralkoxycarbonyl or arylcarbamoyl group or the acyl group of an ⁇ -amino acid optionally N-substituted by a lower alkanoyl or carbamoyl-lower alkanoyl group;
  • R 2 represents cycloalkyl or an optionally substituted aryl or heteroaryl group;
  • R 3 represents hydrogen, alkyl, carbamoyl or an alkanoyl or alkenoyl group optionally substituted by carboxy or esterified or amidated carboxy;
  • R 4 represents an optionally substituted aryl group or an optionally partially saturated heteroaryl group
  • Xi represents methylene, ethylene, a bond, an optionally ketalised carbonyl group or an optionally etherified hydroxymethylene group;
  • X2 represents alkylene, carbonyl or a bond
  • X3 represents carbonyl, oxo-lower alkyl, oxo(aza)-lower alkyl, or an alkyl group optionally substituted by phenyl, hydroxymethyl, optionally esterified or amidated carboxy, or (in other than the ⁇ -position) hydroxy.
  • a particularly preferred compound of formula (VIII) is (2R * , 4S * )-2- benzyl-l-(3,5-dimethylbenzoyl)-N-(4-quinolinylmethyl)-4-piperidineamine; or a pharmaceutically acceptable salt thereof.
  • Another class of NK-1 receptor antagonists of use in the present invention is that described in European Patent Specification No. 0 443 132, i.e. compounds of formula (IX)
  • R 1 is aryl, or a group of the formula:
  • X is CH or N; and Z is O or N-R 5 , in which R 5 is hydrogen or lower alkyl;
  • R 2 is hydroxy or lower alkoxy
  • R 3 is hydrogen or optionally substituted lower alkyl
  • R 4 is optionally substituted ar(lower)alkyl
  • A is carbonyl or sulfonyl; and Y is a bond or lower alkenylene.
  • a particularly preferred compound of formula (IX) is the compound of formula (IXa)
  • NK-1 receptor antagonists of use in the present invention is that described in International Patent Specification No. WO 92/17449, i.e. compounds of the formula (X)
  • R 1 is aryl selected from indanyl, phenyl and naphthyl; heteroaryl selected from thienyl, furyl, pyridyl and quinolyl; and cycloalkyl having 3 to 7 carbon atoms, wherein one of said carbon atoms may optionally be replaced by nitrogen, oxygen or sulfur; wherein each of said aryl and heteroaryl groups may optionally be substituted with one or more substituents, and said C3-7cycloalkyl may optionally be substituted with one or two substituents, said substituents being independently selected from chloro, fluoro, bromo, iodo, nitro, Ci-ioalkyl optionally substituted with from one to three fluoro groups, Ci-ioalkoxy optionally substituted with from one to three fluoro groups, amino, Ci-ioalkyl-S-, C ⁇ -!oalkyl-S(O)-, C ⁇ - ⁇ oalkyl-SO 2 -, phenyl
  • R 2 is thienyl, benzhydryl, naphthyl or phenyl optionally substituted with from one to three substituents independently selected from chloro, bromo, fluoro, iodo, cycloalkoxy having 3 to 7 carbon atoms, Ci-ioalkyl optionally substituted with from one to three fluoro groups and Ci-ioalkoxy optionally substituted with from one to three fluoro groups.
  • a particularly preferred compound of formula (X) is (2S,3S)-3-(2- methoxy-5-trifluoromethoxybenzyl)-amino-2-phenylpiperidine; or a pharmaceutically acceptable salt thereof.
  • Another class of NK-1 receptor antagonists of use in the present invention is that described in International Patent Specification No. WO 95/08549, i.e. compounds of formula (XI) '
  • R i is a C ⁇ . alkoxy group
  • R 2 is
  • R 3 is a hydrogen or halogen atom
  • R 4 and R 5 may each independently represent a hydrogen or halogen atom, or a C ⁇ - 4 alkyl, C ⁇ - 4 alkoxy or trifluoromethyl group
  • R 6 is a hydrogen atom, a C ⁇ - 4 alkyl, (CH2) m cyclopropyl, -S(0) n C ⁇ - 4 alkyl, phenyl, NR 7 R 8 , CH 2 C(O)CF 3 or trifluoromethyl group;
  • R 7 and R 8 may each independently represent a hydrogen atom, or a C ⁇ - 4 alkyl or acyl group; x represents zero or 1; n represents zero, 1 or 2; and m represents zero or 1.
  • Particularly preferred compounds of formula (XI) are (2-methoxy-5- tetrazol-l-yl-benzyl)-([2 ⁇ S,3S]-2-phenyl-piperidin-3-yl)-amine; and [2- methoxy-5-(5-trifluoromethyl-tetrazol-l-yl)-benz3 ]-([2S,3S , ]-2-phenyl- piperidin-3-yl)-amine; or a pharmaceutically acceptable salt thereof.
  • Another class of tachykinin antagonists of use in the present invention is that described in International Patent Specification No. WO 95/14017, i.e. compounds of formula (XII)
  • R is phenyl, 2- or 3-indolyl, 2- or 3-indolinyl, benzothienyl, benzofuranyl, or naphthyl; which R groups may be substituted with one or two halo, C ⁇ - 3 alkoxy, trifluoromethyl, C ⁇ - 4 alkyl, phenyl-C ⁇ -3alkoxy, or C ⁇ - 4 alkanoyl groups;
  • R i is trityl, phenyl, diphenylmethyl, phenoxy, phenylthio, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, indolinyl, indolyl, benzothienyl, hexamethyleneiminyl, benzofuranyl, tetrahydropyridinyl,
  • R x groups may be substituted with halo, C ⁇ - 4 alkyl, C ⁇ - 4 alkoxy, trifluoromethyl, amino, C ⁇ - 4 alkylamino, di(C ⁇ - 4 alkyl)amino, or C 2 - 4 alkanoylamino ; or any one of which R 1 groups may be substituted with phenyl, piperazinyl, C 3 - 8 cycloalkyl, benzyl, C ⁇ -4alkyl, piperidinyl, pyridinyl, pyrimidinyl, C 2 -Galkanoylamino, pyrrolidinyl, d-ealkanoyl, or C ⁇ - 4 alkoxycarbonyl; any one of which groups may be substituted with halo, d.
  • R 5 is pyridyl, anilino-(C ⁇ - 3 alkyl)-, or anilinocarbonyl;
  • R 2 is hydrogen, d-4alkyl, C ⁇ -4alkylsulfonyl, carboxy-(C ⁇ - 3 alkyl)-, Ci-salkoxycarbonyMCi-salkyi)-, or -CO-R 6 ;
  • R 6 is hydrogen, C ⁇ -4alkyl, C ⁇ -3haloalkyl, phenyl, C ⁇ - 3 alkoxy, C ⁇ - 3 hydroxyalkyl, amino, C ⁇ - 4 alkylamino, di(C ⁇ - 4 alkyl)amino, or -(CH 2 ) q -R 7 ; q is zero to 3;
  • R 7 is carboxy, C ⁇ - 4 alkoxycarbonyl, C ⁇ . 4 alkylcarbonyloxy, amino, C ⁇ - alkylamino, di(C ⁇ - alkyl)amino, Ci-ealkoxycarbonylamino, or phenoxy, phenylthio, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, indolinyl, indolyl, benzothienyl, benzofuranyl, quinolinyl, phenyl-(C ⁇ - 4 alkyl)-, quinolinyl- (C ⁇ .
  • R 7 groups may be substituted with phenyl, piperazinyl, C 3 - 8 cycloalkyl, benzyl, piperidinyl, pyridinyl, pyrimidinyl, pyrrolidinyl, d ealkanoyl, or C ⁇ - 4 alkoxycarbonyl; any of which groups may be substituted with halo, trifluoromethyl, amino, d- 4 alkoxy, C ⁇ - 4 alkyl, d- 4 alkylamino, di(C ⁇ -4alkyl)amino, or C 2 . alkanoylamino ;
  • R 8 is hydrogen or Ci-ealkyl
  • R 3 is phenyl, phenyl-(C ⁇ -6alkyl)-, C 3 -8cycloalkyl, Cs-scycloalkenyl,
  • R 4 is hydrogen or C ⁇ -3alkyl; with the proviso that if R i is hydrogen or halo, R 3 is phenyl, phenyl-(d-6alkyl)-, C3-8cycloalkyl, Cs-scycloalkenyl, or naphthyl.
  • a particularly preferred compound of formula (XII) is [N-(2- methoxybenzyl)acetylamino]-3-(lH-indol-3-yl)-2-[N-(2-(4-piperidin-l- yl)piperidin-l-yl)acetylamino]propane; or a pharmaceutically acceptable salt thereof.
  • the preferred stereochemistry of the ⁇ -carbon is either (R) when the substituent is an alkyl (e.g. methyl) group or (S) when the substituent is a hydroxyalkyl (e.g. hydroxymethyl) group.
  • suitable alkyl groups include straight-chained and branched alkyl groups containing from 1 to 6 carbon atoms. Typical examples include methyl and ethyl groups, and straight - chained or branched propyl and butyl groups. Particular alkyl groups are methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl and tert-butyl.
  • suitable alkenyl groups include straight-chained and branched alkenyl groups containing from 2 to 6 carbon atoms. Typical examples include vinyl and allyl groups.
  • suitable alkynyl groups include straight-chained and branched alkynyl groups containing from 2 to 6 - carbon atoms. Typical examples include ethynyl and propargyl groups.
  • suitable cycloalkyl groups include groups containing from 3 to 7 carbon atoms. Particular cycloalkyl groups are cyclopropyl and cyclohexyl. Unless otherwise defined herein, suitable aryl groups include phenyl and naphthyl groups.
  • a particular aryl-C 1 6 alkyl, e.g. phenyl-Ci-ealkyl, group is benzyl.
  • suitable heteroaryl groups include pyridyl, quinolyl, isoquinolyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyranyl, furyl, benzofuryl, thienyl, benzthienyl, imidazolyl, oxadiazolyl and thiadiazolyl groups.
  • halogen as used herein includes fluorine, chlorine, bromine and iodine.
  • the compounds of use in this invention may have one or more asymmetric centres and can therefore exist as enantiomers and possibly as diastereoisomers. It is to be understood that the present invention relates to the use of all such isomers and mixtures thereof.
  • Suitable pharmaceutically acceptable salts of the NK-1 receptor antagonists of use in the present invention include acid addition salts which may, for example, be formed by mixing a solution of the compound with a solution of a pharmaceutically acceptable non-toxic acid such as hydrochloric acid, fumaric acid, maleic acid, succinic acid, acetic acid, citric acid, tartaric acid, carbonic acid, phosphoric acid or sulphuric acid.
  • Salts of amine groups may also comprise the quaternary ammonium salts in which the amino nitrogen atom carries an alkyl, alkenyl, alkynyl or aralkyl group.
  • the present invention also contemplates salts thereof, preferably non-toxic pharmaceutically acceptable salts thereof, such as the sodium, potassium and calcium salts thereof.
  • Suitable pharmaceutically acceptable salts of the anorectic agent of use in the combinations of the present invention include those salts described above in relation to the salts of NK-1 receptor antagonists.
  • the present invention accordingly provides the use of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII), for the manufacture of a medicament for the treatment or prevention of eating disorders.
  • the present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII).
  • a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII).
  • a pharmaceutical composition for the treatment or prevention of eating disorders comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII), together with at least one pharmaceutically acceptable carrier or excipient.
  • a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII), together with at least one pharmaceutically acceptable carrier or excipient.
  • the NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) may be used in combination with an anorectic agent, present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of eating disorders.
  • Such combined preparations may be, for example, in the form of a twin pack.
  • a product comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
  • the present invention accordingly provides the use of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a - halogenated amphetamine derivative for the manufacture of a medicament for the treatment or prevention of eating disorders.
  • a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a - halogenated amphetamine derivative for the manufacture of a medicament for the treatment or prevention of eating disorders.
  • the present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative such that together they give effective relief.
  • a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative such that together they give effective relief.
  • a pharmaceutical composition comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative together with at least one pharmaceutically acceptable carrier or excipient.
  • a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative together with at least one pharmaceutically acceptable carrier or excipient.
  • a product comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
  • a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
  • halogenated amphetamine derivatives are selected from the group consisting of fenfluramine and dexfenfluramine.
  • the present invention accordingly provides the use of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, for the manufacture of a medicament for the treatment or prevention of eating disorders.
  • the present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and an amount of a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, such that together they give effective relief.
  • a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and an amount of a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, such that together they give effective relief.
  • a pharmaceutical composition comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, together with at least one pharmaceutically acceptable carrier or excipient.
  • a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, together with at least one pharmaceutically acceptable carrier or excipient.
  • a product comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
  • a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
  • the NK-1 receptor antagonist and the anorectic agent may be formulated in a single pharmaceutical composition or alternatively in individual pharmaceutical compositions for simultaneous, separate or sequential use in accordance with the present invention.
  • the compositions according to the present invention are in unit dosage forms such as tablets, pills, capsules, powders, granules, solutions or suspensions, or suppositories, for oral, parenteral or rectal administration, by inhalation or insufflation or administration by trans - dermal patches or by buccal cavity absorption wafers.
  • Oral dosage forms are particularly preferred (e.g. tablets, capsules, pills or wafers).
  • the principal active ingredient is mixed with a pharmaceutical carrier, e.g.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a non-toxic pharmaceutically acceptable salt thereof.
  • preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention.
  • the tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
  • liquid forms in which the novel compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavoured syrups, aqueous or oil suspensions, and flavoured emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, peanut oil or soybean oil, as well as elixirs and similar pharmaceutical vehicles.
  • Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone or gelatin.
  • compositions for administration by injection include those comprising a NK-1 receptor antagonist as the active ingredient, in association with a surface-active agent (or wetting agent or surfactant) or in the form of an emulsion (as a water-in-oil or oil-in-water emulsion).
  • a surface-active agent or wetting agent or surfactant
  • Suitable surface-active agents include, in particular, non-ionic agents, such as polyoxyethylenesorbitans (e.g. TweenTM 20, 40, 60, 80 or 85) and other sorbitans (e.g. SpanTM 20, 40, 60, 80 or 85).
  • Compositions with a surface -active agent will conveniently comprise between 0.05 and 5% surface-active agent, and preferably between 0.1 and 2.5%. It will be appreciated that other ingredients may be added, for example mannitol or other pharmaceutically acceptable vehicles, if necessary.
  • Suitable emulsions may be prepared using commercially available fat emulsions, such as IntralipidTM, LiposynTM, InfonutrolTM, ipofundinTM and LipiphysanTM.
  • the active ingredient may be either dissolved in a pre- mixed emulsion composition or alternatively it may be dissolved in an oil (e.g. soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g. egg phospholipids, soybean phospholipids or soybean lecithin) and water.
  • an oil e.g. soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil
  • a phospholipid e.g. egg phospholipids, soybean phospholipids or soybean lecithin
  • other ingredients may be added, for example glycerol or glucose, to adjust the tonicity of the emulsion.
  • Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%.
  • the fat emulsion will preferably comprise fat droplets between 0.1 and l.O ⁇ m, particularly 0.1 and 0.5 ⁇ m, and have a pH in the range of 5.5 to 8.0.
  • Particularly preferred emulsion compositions are those prepared by mixing a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) with IntralipidTM or the components thereof (soybean oil, egg phospholipids, glycerol and water).
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulised by use of inert gases. Nebulised solutions may be breathed directly from the nebulising device or the nebulising device may be attached to a face mask, tent or intermittent positive pressure breathing machine.
  • Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
  • compositions of the present invention may also be presented for administration in the form of trans-dermal patches using conventional technology.
  • the compositions may also be administered via the buccal cavity using, for example, absorption wafers.
  • the present invention further provides a process for the preparation of a pharmaceutical composition comprising a NK-1 receptor antagonist and an anorectic agent, which process comprises bringing a NK-1 receptor antagonist and an anorectic agent, into association with a pharmaceutically acceptable carrier or excipient.
  • the NK-1 receptor antagonist and an anorectic agent When administered in combination, either as a single or as separate pharmaceutical composition(s), the NK-1 receptor antagonist and an anorectic agent, are presented in a ratio which is consistent with the manifestation of the desired effect.
  • the ratio by weight of the NK-1 receptor antagonist and the anorectic agent will suitably be between 0.001 to 1 and 1000 to 1, and especially between 0.01 to 1 and 100 to 1.
  • a suitable dosage level for the NK-1 receptor antagonist about 0.05 to 1500mg per day, preferably about 0.25 to 1500mg per day, and especially about 0.25 to 500mg/kg per day.
  • the compounds may be administered on a regimen of up to 6 times per day, preferably 1 to 4 times per day, especially 1 or 2 times daily.
  • a suitable dosage level for the anorectic agent is about 0.5 to 1500mg per day, preferably about 2.5 to lOOOmg per day, and especially about 2.5 to 500mg per day.
  • the compounds may be administered on a regimen of up to 6 times per day, preferably 1 to 4 times per day, especially 1 or 2 times daily.
  • the amount of the NK-1 receptor antagonist and (where present) the anorectic agent required for use in the treatment or prevention of eating disorders will vary not only with the particular compounds or compositions selected but also with the route of administration, the nature of the condition being treated, and the age and condition of the patient, and will ultimately be at the discretion of the patient's physician or pharmacist.
  • the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) may be prepared by the methods described in EP-A-0 577 394 (or WO 95/16679), WO 95/18124, WO 95/23798, WO 96/05181, EP-A-0 436 334, WO 93/21155, EP-A-0 591 040, EP-A-0 532 456, EP-A-0 443 132, WO 92/17449, WO 95/08549 and WO 95/14017, respectively.
  • NK-1 receptor antagonists of the formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) for use in the present invention are compounds which are potent NK-1 receptor antagonists, i.e. compounds with an NK-1 receptor affinity (ICso) of less than lOOnM.
  • ICso NK-1 receptor affinity
  • IC50 NK-1 receptor affinity
  • Especially preferred NK-1 receptor antagonists of use in the present invention are orally active, long acting, CNS-penetrant NK-1 receptor antagonists, identified using a combination of the following assays:
  • NK-1 Receptor binding NK-1 receptor binding assays are performed in intact Chinese hamster ovary (CHO) cells expressing the human NK-1 receptor using a modification of the assay conditions described by Cascieri et al, J. Pharmacol. Exp. Ther., 1992, 42, 458. Typically, the receptor is expressed at a level of 3xl0 5 receptors per cell. Cells are grown in monolayer culture, detached from the plate with enzyme-free dissociation solution (Speciality Media Inc.), and washed prior to use in the assay.
  • I-Tyr 8 -substance P (O.lnM, 2000Ci/mmol; New England Nuclear) is incubated in the presence or absence of test compounds (dissolved in 5 ⁇ l dimethylsulphoxide, DMSO) with 5xl0 4 CHO cells.
  • Ligand binding is performed in 0.25ml of 50mM Tris-HCl, pH7.5, containing 5mM MnCl 2 , 150mM NaCl, 0.02% bovine serum albumin (Sigma), 50 ⁇ g/ml chymostatin (Peninsula), O.lnM phenylmethylsulphonyl fluoride, 2 ⁇ g/ml pepstatin, 2 ⁇ g/ml leupeptin and 2.8 ⁇ g/ml furoyl saccharine.
  • the incubation proceeds at room temperature until equilibrium is achieved (>40 minutes) and the receptor-ligand complex is harvested by filtration over GF/C filters pre-soaked in 0.1% polyethylenimine using a Tomtek 96-well harvester. Non-specific binding is determined using excess substance P (l ⁇ M) and represents ⁇ 10% of total binding.
  • ASSAY 2 Gerbil Foot-Tapping
  • CNS-penetrant NK-1 receptor antagonists for use in the present invention can be identified by their ability to inhibit foot tapping in gerbils induced by anxiogenic agents (such as pentagastrin) or central infusion of NK-1 receptor agonists such as GR73632, or caused by aversive stimulation such as foot shock or single housing, based on the method of Rupniak & Williams, Eur. J. Pharmacol, 1994, 265, 179.
  • test compounds Male or female Mongolian gerbils (35-70g) are anaesthetised by inhalation of an isoflurane/oxygen mixture to permit exposure of the jugular vein in order to permit administration of test compounds or vehicle in an injection volume of approximately 5ml/kg i.v.
  • test compounds may be administered orally or by subcutaneous or intraperitoneal routes. A skin incision is then made in the midline of the scalp to expose the skull.
  • An anxiogenic agent e.g. pentagastrin
  • a selective NK-1 receptor agonist e.g.
  • GR73632 (d Ala[L-Pro 9 ,Me-Leu 10 ]- substance P-(7-ll)) is infused directly into the cerebral ventricles (e.g. 3pmol in 5 ⁇ l i.c.v., depending on test substance) by vertical insertion of a cuffed 27 gauge needle to a depth of 4.5mm below bregma.
  • the scalp incision is closed and the animal allowed to recover from anaesthesia in a clear perspex observation box (approximately 25cm x 20cm x 20cm).
  • the duration and/or intensity of hind foot tapping is then recorded continuously for approximately 5 minutes.
  • the ability of test compounds to inhibit foot tapping evoked by aversive stimulation such as foot shock or single housing, may be studied using a similar method of quantification.
  • ferrets Individually housed male ferrets (1.0 -2.5 kg) are dosed orally by gavage with test compound. Ten minutes later they are fed with approximately lOOg of tinned cat food. At 60 minutes following oral dosing, cisplatin (lOmg/kg) is given i.v. via a jugular vein catheter inserted under a brief period of halothane anaesthesia. The catheter is then removed, the jugular vein ligated and the skin incision closed. The ferrets recover rapidly from the anaesthetic and are mobile within 10-20 minutes. The animals are observed continuously during recovery from the anaesthetic and for 4 hours following the cisplatin injection, after which time the animals are killed humanely. The numbers of retches and vomits occurring during the 4 hours after cisplatin administration are recorded by trained observers.
  • ASSAY 4 Separation-Induced Vocalisation
  • mice Male and female guinea-pigs pups are housed in family groups with their mothers and littermates throughout the study. Experiments are commenced after weaning when the pups are at least 2 weeks old. Before entering an experiment, the pups may be screened to ensure that a vigorous vocalisation response is reproducibly elicited following maternal separation. The pups are placed individually in an observation cage (approximately 55cm x 39cm x 19cm) in a room physically isolated from the home cage for approximately 15 minutes and the duration and or number of vocalisation during this baseline period is recorded. Those animals which vocalise for longer than 5 minutes are employed for drug challenge studies (approximately 50% of available pups may fail to reach this criterion).
  • each pup On test days each pup receives an oral dose or an s.c. or i.p. injection of test compound or vehicle and is then immediately returned to the home cage with its mother and siblings for at least 30 to 60 minutes (or for up to 4 hours following an oral dose, dependent upon the oral pharmacokinetics of the test compound) before social isolation for 15 minutes as described above.
  • the duration and/or number of vocalisation on drug treatment days may be expressed as a percentage of the pre- treatment baseline value for each animal or compared with values obtained in vehicle-treated animals. The same subjects may be retested once weekly for up to 6 weeks. Between 6 and 8 animals receive each test compound at each dose tested.
  • a suitable selection cascade for NKi antagonists of use according to the present invention is as follows: (i) Determine affinity for human NKi receptor in radioligand binding studies (Assay 1); select compounds with ICso ⁇ lOnM, preferably IC50 ⁇ 2nM, especially IC50 ⁇ InM.
  • step (iv) Determine oral bioavailability of compounds by pharmacokinetic analysis, activity in gerbil foot tapping assay following oral administration and/or by ability to inhibit cisplatin-induced emesis in ferrets (Assay 3); select compounds with ID90 ⁇ 3mg/kg p.o., and preferably Particularly preferred compounds of use in the present invention are identified using steps (i) to (iv) followed by step (v):
  • Yet further preferred compounds of use in the present invention may be selected from those compounds which satisfy the NK-1 receptor binding criteria of step (i) which, in addition, have ⁇ 5-fold shift in affinity when incubated in the presence of human serum albumin (HSA) to show non-specific protein binding.
  • HSA human serum albumin
  • a NK-1 receptor antagonist of use in the present invention is the compound 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)- ethoxy)-3-(S)-(4-fluorophenyl)-4-(3-(5-oxo-lH,4H-l,2,4-triazolo)methyl)- morpholine, the preparation of which is described in International Patent Specification No. WO 95/16679. In the aforementioned assays, this compound has the following activity:
  • a NK-1 receptor antagonist of use in the present invention is the compound 2-(R)-(l-(R)-(3,5- bis(trifluoromethyl)phenyl)ethoxy)-4-(5-(N,N-dimethylamino)methyl-l,2,3- triazol-4-yl)methyl-3-(S)-phenylmorpholine, the preparation of which is described in International Patent Specification No. WO 95/18124. In the aforementioned assays, this compound has the following activity:
  • the following assay has been used to demonstrate the potentiation of the anorectic effects of dexfenfluramine in diet-induced obese mice when co-administered with a NK-1 receptor antagonist.
  • mice Male C57BL/J mice were obtained from Jackson Labs at 3 weeks of age. Half the mice were maintained on a wet diet consisting of sweetened condensed milk and standard ground rodent chow (70%:30%, vol:vol). Fresh wet chow was provided daily. These mice will be referred to as diet- induced obese (DIO). The other half was maintained on just ground rodent chow. These will be referred to as Non-Obese Littermates (NOL). Both food and water were supplied ad libitum. Mice were housed with a 12 hour light/dark cycle (4.00am lights on) through out the course of the described studies.
  • DIO diet- induced obese mice
  • NOL Non-Obese Littermates
  • mice were weighed bi-weekly until a point that both DIO and NOL mice were weight stable (approximately 20 weeks). At this time, DIO mice weighed significantly more than NOL mice (p 2 0.01). DIO mice also exhibited elevated insulin and glucose levels, as well as polyuria.
  • mice received two injections approximately 30 mins apart. All injections were administered ip., in a volume of 0.2 ml between 3.00pm and 3.30pm. Fresh chow was provided at the time of injection. Food intake was measured 16 hours post-injection for each mouse.
  • Results shown in Figure 1 are expressed as inhibition of food intake relative to that of saline treated animals.
  • mice are treated with dexfenfluramine, GR 205171, or combinations of dexfenfluramine with decreasing doses of GR 205171, similar to those used in the evaluation of food intake. Mice are dosed once daily, for 7 days with body weights being measured at the start and conclusion of the study.
  • formulations may be prepared with separate active ingredients or with a combination of active ingredients in one composition.
  • the ratio of the NK-1 receptor antagonist and the anorectic agent will depend upon the choice of active ingredients.
  • the active ingredients cellulose, lactose and a portion of the corn starch are mixed and granulated with 10% corn starch paste.
  • the resulting granulation is sieved, dried and blended with the remainder of the corn starch and the magnesium stearate.
  • the resulting granulation is then compressed into tablets containing 50mg, lOOmg and 300mg of the NK-1 receptor antagonist per tablet.
  • the sodium phosphate, citric acid monohydrate and sodium chloride are dissolved in a portion of the water.
  • the active ingredients are dissolved or suspended in the solution and made up to volume.

Abstract

The present invention provides the use of an NK-1 receptor antagonist, optionally with an anorectic agent, for the manufacture of a medicament for the treatment or prevention of eating disorders, methods of treatment using the NK-1 receptor antagonist and pharmaceutical compositions and products containing it.

Description

USE OF NK-1 RECEPTOR ANTAGONISTS FOR TREATING EATING DISORDERS
This invention relates to the treatment or prevention of eating disorders by the administration of a NK-1 receptor antagonist, optionally in combination with an anorectic agent.
Eating disorders commonly arise through an imbalance in a subject's appetite, or desire to eat. It is recognised that appetite is influenced by the interaction of central and peripheral systems, most likely acting through the effects of neuropeptides on the so-called feeding and saiety centres in the hypothalamus region of the brain. For instance, neuropeptides released by the gut in response to a meal may serve to modulate the intake of further food.
Alterations of appetite may lead to eating disorders including obesity, bulimia nervosa, and compulsive eating disorders.
Obesity is a chronic disease that is highly prevalent in modern society and is associated not only with a social stigma, but also with decreased life span and numerous medical problems, including adverse psychological development, reproductive disorders such as polycystic ovarian disease, dermatological disorders such as infections, varicose veins, Acanthosis nigricans, and eczema, exercise intolerance, diabetes mellitus, insulin resistance, hypertension, hypercholesterolemia, cholelithiasis, osteoarthritis, orthopedic injury, thromboembolic disease, cancer, and coronary heart disease. Rissanen et al, British Medical Journal, 301:835-837 (1990).
Bulimia nervosa is characterised by recurrent episodes of overeating, or binges, followed by severe dieting often associated with self- induced vomiting, abuse of laxatives or diuretics, or excessive exercise to avoid weight gain. Frequent vomiting or purging may result in electrolyte disturbances and erosion of dental enamel. Complusive eating disorders may or may not be associated with other neurological disorders. One well characterised compulsive eating disorder is Prader-Willi syndrome, a congential disorder characterised by infantile hypotonia, hypogonadism and facial dysmorphism, with subsequent development of hyperplagia and abnormalities of behaviour and intellect.
Treatment regimens for eating disorders typically include the use of anorectic agents, such as amphetamine derivatives. jo-Chloroamphetamine and other halogenated amphetamines promote serotonin (5-hydroxytryptamine; 5-HT) release from platelets and neurons. A rapid release of serotonin is followed by a prolonged and selective depletion of serotonin in the brain. The most widely used example of this class of compound is fenfluramine and its (S)-isomer, dexfenfluramine. The precise mechanism of action of these compounds is uncertain, however, fenfluramine and dexfenfluramine are both useful in the treatment of bulimia nervosa and obesity, and fenfluramine has also produced promising results in the management of Prader-Willi syndrome.
Neurokinin 1 (NK-1; substance P) receptor antagonists are being developed for the treatment of a number of physiological disorders associated with an excess or imbalance of tachykinins, and in particular substance P. Examples of such conditions include disorders of the central nervous system such as anxiety, depression and psychosis (see, for instance, International (PCT) patent specification Nos. WO 95/16679, WO 95/18124 and WO 95/23798). We have now found that NK-1 receptor antagonists are effective in the treatment of eating disorders, as evidenced by their activity in vivo in a model of diet-induced obesity.
Furthermore, a combination of an anorectic agent with a NK-1 receptor antagonist may provide an enhanced anorectic effect. They may also provide for a rapid onset of action to combat eating disorders thereby enabling prescription on an "as-needed" basis. A particularly preferred class of NK-1 receptor antagonists of use in the present invention are those which are able to cross the blood-brain barrier, otherwise known as CNS- or brain-penetrant compounds. The present invention accordingly provides the use of a NK-1 receptor antagonist for the manufacture of a medicament for the treatment or prevention of eating disorders.
The present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist.
In a further aspect of the present invention, there is provided a pharmaceutical composition for the treatment or prevention of eating disorders comprising a NK-1 receptor antagonist, together with at least one pharmaceutically acceptable carrier or excipient. In a further embodiment of the present invention there is provided the use of a NK-1 receptor antagonist for the manufacture of a medicament for the treatment or prevention of obesity.
The present invention also provides a method for the treatment or prevention of obesity, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist.
In an alternative embodiment of the present invention there is provided the use of a NK-1 receptor antagonist for the manufacture of a medicament for the treatment or prevention of bulimia nervosa. The present invention also provides a method for the treatment or prevention of bulimia nervosa, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist.
In a further embodiment of the present invention there is provided the use of a NK-1 receptor antagonist for the manufacture of a medicament for the treatment or prevention of compulsive eating disorders.
The present invention also provides a method for the treatment or prevention of compulsive eating disorders, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist.
In an alternative embodiment of the present invention there is provided the use of a NK-1 receptor antagonist for the manufacture of a medicament for reducing the total body fat mass in an obese mammal, especially a human.
The present invention also provides a method for reducing the total body fat mass in an obese mammal, especially a human, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist. The present invention further provides the use of a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for the treatment or prevention of eating disorders.
The present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
In a further aspect of the present invention, there is provided a pharmaceutical composition comprising a NK-1 receptor antagonist and an anorectic agent, together with at least one pharmaceutically acceptable carrier or excipient.
It will be appreciated that the NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of eating disorders. Such combined preparations may be, for example, in the form of a twin pack. In a further or alternative aspect of the present invention, there is therefore provided a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders. In a further embodiment of the present invention there is provided the use of a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for the treatment or prevention of obesity. The present invention also provides a method for the treatment or prevention of obesity, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
It will be appreciated that the NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of obesity. Such combined preparations may be, for example, in the form of a twin pack.
In a further or alternative aspect of the present invention, there is therefore provided a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of obesity.
In an alternative embodiment of the present invention there is provided the use of a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for the treatment or prevention of bulimia nervosa. The present invention also provides a method for the treatment or prevention of bulimia nervosa, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief. It will be appreciated that the NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of bulimia nervosa. Such combined preparations may be, for example, in the form of a twin pack.
In a further or alternative aspect of the present invention, there is therefore provided a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of bulimia nervosa.
In a further embodiment of the present invention there is provided the use of a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for the treatment or prevention of compulsive eating disorders.
The present invention also provides a method for the treatment or prevention of compulsive eating disorders, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
It will be appreciated that the NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of compulsive eating disorders. Such combined preparations may be, for example, in the form of a twin pack.
In a further or alternative aspect of the present invention, there is therefore provided a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of compulsive eating disorders.
In an alternative embodiment of the present invention there is provided the use of a NK-1 receptor antagonist and an anorectic agent for the manufacture of a medicament for reducing the total body fat mass in an obese mammal, especially a human. The present invention also provides a method for reducing the total body fat mass in an obese mammal, especially a human, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
It will be appreciated that the NK-1 receptor antagonist and anorectic agent may be present as a combined preparation for simultaneous, separate or sequential use for reducing the total body fat mass in an obese mammal, especially a human. Such combined preparations may be, for example, in the form of a twin pack.
In a further or alternative aspect of the present invention, there is therefore provided a product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in reducing the total body fat mass in an obese mammal, especially a human.
It will be appreciated that when using a combination of the present invention, both the NK-1 receptor antagonist and the anorectic agent will be administered to a patient, within a reasonable period of time. The compounds may be in the same pharmaceutically acceptable carrier and therefore administered simultaneously. They may be in separate pharmaceutical carriers such as conventional oral dosage forms which are taken simultaneously. The term "combination" also refers to the case where the compounds are provided in separate dosage forms and are administered sequentially. Therefore, by way of example, the anorectic agent may be administered as a tablet and then, within a reasonable period of time, the NK-1 receptor antagonist may be administered either as an oral dosage form such as a tablet or a fast-dissolving oral dosage form. By a "fast dissolving oral formulation" is meant, an oral delivery form which when placed on the tongue of a patient, dissolves within about 10 seconds. By "reasonable period of time" is meant a time period that is not in excess of about 1 hour. That is, for example, if the anorectic agent is provided as a tablet, then within one hour, the NK-1 receptor antagonist should be administered, either in the same type of dosage form, or another dosage form which provides effective delivery of the medicament. The compositions of the present invention are useful for the prevention or treatment of eating disorders. As used herein, the term "eating disorders" includes obesity, bulimia nervosa and compulsive eating disorders. As used herein "obesity" refers to a condition whereby a mammal has a Body Mass Index (BMI), which is calculated as weight per height squared (kg/m2), of at least 25.9. Conventionally, those persons with normal weight, have a BMI of 19.9 to less than 25.9.
The obesity herein may be due to any cause, whether genetic or environmental. Examples of disorders that may result in obesity or be the cause of obesity include overeating and bulimia, polycystic ovarian disease, craniopharyngioma, the Prader-Willi Syndrome, Frohlich's syndrome, Type II diabetes, GH-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g, children with acute lymphoblastic leukemia.
"Treatment" refers to reducing the BMI of the mammal to less than about 25.9, and maintaining that weight for at least 6 months. The treatment suitably results in a reduction in food or calorie intake by the mammal.
"Prevention" refers to preventing obesity from occurring if the treatment is administered prior to the onset of the obese condition. Moreover, if treatment is commenced in already obese subjects, such treatment is expected to prevent, or to prevent the progression of, the medical sequelae of obesity, such as, e.g., arteriosclerosis, Type II diabetes, polycycstic ovarian disease, cardiovascular diseases, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, and cholelithiasis.
Thus, in one aspect, this invention relates to the inhibition and/or complete suppression of lipogenesis in obese mammals, i.e., the excessive accumulation of lipids in fat cells, which is one of the major features of human and animal obesity, as well as loss of total body weight. In another aspect, the invention ameliorates the conditions that are a consequence of the disease, such as preventing or arresting the progression of polycystic ovarian disease so that the patient is no longer infertile, and increasing the insulin sensitivity and/or decreasing or eliminating the need or usage of insulin in a diabetic patient, e.g., one with adult-onset diabetes or Type II diabetes.
"Mammals" include animals of economic importance such as bovine, ovine, and porcine animals, especially those that produce meat, as well as domestic animals, sports animals, zoo animals, and humans, the latter being preferred.
The compositions of the present invention are especially useful for the treatment of or prevention of eating disorders where the use of an anorectic agent is generally prescribed. By the use of a combination of a NK-1 receptor antagonist and an anorectic agent in accordance with the present invention, it is now also possible to treat or prevent eating disorders in patients for whom conventional anorectic therapy might not be wholly successful or where dependance upon the anorectic therapy is prevalent.
Suitable anoretic agents of use in the combinations of the present invention include, but are not limited to, aminorex, amphechloral, amphetamine, benzphetamine, chlorphentermine, clobenzorex, cloforex, clominorex, clortermine, cyclexedrine, dexfenfluramine, dextroamphetamine, diethylpropion, diphemethoxidine, N- ethylamphetamine, fenbutrazate, fenfluramine, fenisorex, fenproporex, fludorex, fluminorex, furfurylmethylamphetamine, levamfetamine, levophacetoperane, mazindol, mefenorex, metamfepramone, methamphetamine, norpseudoephedrine, pentorex, phendimetrazine, phenmetrazine, phentermine, phenylpropanolamine, picilorex and sibutramine; and pharmaceutically acceptable salts thereof.
Particularly preferred anorectic agents include amphetamine and derivatives thereof such as amphetamine, benzphetamine, chlorphentermine, clobenzorex, cloforex, clotermine, dexfenfluramine, dextroamphetamine, diethylpropion, N-ethylamphetamine, fenfluramine, fenproporex, furfurylmethylamphetamine, levamfetamine, mefenorex, metamfepramone, methamphetamine, norpseudoephedrine, pentorex, phendimetrazine, phenmetrazine, phentermine, phenylpropanolamine, picilorex and sibutramine; and pharmaceutically acceptable salts thereof.
A particularly suitable class of anorectic agent are the halogenated amphetamine derivatives, including chlorphentermine, cloforex, clortermine, dexfenfluramine, fenfluramine, picilorex and sibutramine; and pharmaceutically acceptble salts thereof;
Particularly preferred halogenated amphetamine derivatives of use in the present invention include: fenfluramine and dexfenfluramine, and pharmaceutically acceptable salts thereof.
Another particularly preferred anorectic agent is phentermine.
NK-1 receptor antagonists of use in the present invention are described in published European Patent Specification Nos. 0 360 390, 0 394 989, 0 429 366, 0 443 132, 0 482 539, 0 512 901, 0 512 902, 0 514 273, 0 514 275, 0 517 589, 0 520 555, 0 522 808, 0 528 495, 0 532 456, 0 533 280, 0 536 817, 0 545 478, 0 577 394, 0 590 152, 0 599 538, 0 610 793, 0 634 402, 0 686 629, 0 693 489, 0 694 535, 0 699 655, 0 699 674, 0 707 006, 0 708 101, 0 714 891, 0 723 959, 0 733 632 and 0 776 893; and in International Patent Specification Nos. 90/05525, 90/05729, 91/09844, 91/18899, 92/01688, 92/06079, 92/12151, 92/15585, 92/17449, 92/20661, 92/20676, 92/21677, 93/00330, 93/00331, 93/01159, 93/01165, 93/01169, 93/01170, 93/06099, 93/09116, 93/10073, 93/14113, 93/18023, 93/19064, 93/21155, 9321181, 93/23380, 93/24465, 94/01402, 94/02461, 94/03429, 94/03445, 94/04494, 94/04496, 94/05625, 94/07843, 94/10165, 94/10167, 94/10168, 94/10170, 94/11368, 94/13639, 94/13663, 94/14767, 94/15903, 94/19320, 94/19323, 94/20500, 94/26735, 94/26740, 94/29309, 95/02595, 95/04040, 95/04042, 95/06645, 95/07886, 95/07908, 95/08549, 95/11880, 95/14017, 95/15311, 95/16679, 95/17382, 95/18124, 95/18129, 95/19344, 95/20575, 95/21819, 96/22525, 95/23798, 95/26338, 95/28418, 95/30674, 95/30687, 96/05193, 96/05203, 96/06094, 96/07649, 96/10562, 96/16939, 96/18643, 96/20197, 96/21661, 96/29304, 96/29317, 96/29326, 96/29328, 96/31214, 96/32385, 96/37489, 97/01553, 97/01554, 97/03066, 97/08144, 97/14671, 97/17362, 97/18206, 97/19084, 97/19942 and 97/21702; and in British Patent Specification Nos. 2 266 529, 2 268 931, 2 269 170, 2 269 590, 2 271 774, 2 292 144, 2 293 168, 2 293 169, and 2 302 689.
Particularly preferred NK-1 receptor antagonists are those described in European Patent Specification No. 0 577 394, i.e. compounds of formula (I):
or a pharmaceutically acceptable salt thereof, wherein:
R1 is selected from the group consisting of:
(1) hydrogen;
(2) Ci-ealkyl, unsubstituted or substituted with one or more of the substituents selected from:
(a) hydroxy,
(b) oxo,
(c) Ci-ealkoxy, (d) phenyl-Cι-3alkoxy,
(e) phenyl,
(f) -CN,
(g) halo, (h) -NR9Ri°, wherein R9 and R10 are independently selected from:
(i) hydrogen, (ii) Ci-βalkyl, (iii) hydroxy-Ci-6alkyl, and (iv) phenyl,
(i) -NR9CORi°, wherein R9 and Rio are as defined above,
(j) -NR9CO2R10, wherein R9 and R10 are as defined above,
(k) -CONR9R °, wherein R9 and Rio are as defined above,
(1) -COR9, wherein R9 is as defined above, (m) -CO2R9, wherein R9 is as defined above,
(n) heterocycle, wherein the heterocycle is selected from the group consisting of:
(A) benzimidazolyl,
(B) benzofuranyl, (C) benzthiophenyl,
(D) benzoxazolyl,
(E) furanyl,
(F) imidazolyl,
(G) indolyl, (H) isoxazolyl,
(I) isothiazolyl,
(J) oxadiazolyl,
(K) oxazolyl,
(L) pyrazinyl, (M) pyrazolyl,
(N) pyridyl, (O) pyrimidyl,
(P) pyrrolyl,
(Q) quinolyl,
(R) tetrazolyl, (S) thiadiazolyl,
(T) thiazolyl,
(U) thienyl,
(V) triazolyl,
(W) azetidinyl, (X) 1,4-dioxanyl,
(Y) hexahydroazepinyl,
(Z) oxanyl,
(AA) piperazinyl,
(AB) piperidinyl, (AC) pyrrolidinyl,
(AD) tetrahydrofuranyl, and
(AE) tetrahydrothienyl, and wherein the heterocylcle is unsubstituted or substituted with one or more substituent(s) selected from: (i) Ci-ealkyl, unsubstituted or substituted with halo, -CF3,
-OCH3, or phenyl,
(ii) Ci-βalkoxy,
(iii) oxo,
(iv) hydroxy, (v) thioxo,
(vi) -SR9, wherein R9 is as defined above,
(vii) halo,
(viii) cyano,
(ix) phenyl, (x) trifluoromethyl, (xi) -(CH2)m-NR9Ri°, wherein m is 0, 1 or 2, and R9 and R10 are as defined above,
(xii) -NR9CORi°, wherein R9 and Rio are as defined above, (xiii) -CONR9Ri°, wherein R9 and R10 are as defined above, (xiv) -CO2R9, wherein R9 is as defined above, and
(xv) -(CH2)m-OR9, wherein m and R9 are as defined above;
(3) C2-6alkenyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy, (b) oxo,
(c) d-βalkoxy,
(d) phenyl-Cι-3alkoxy,
(e) phenyl,
(f) -CN, (g) halo,
(h) -CONR9Ri°, wherein R9 and R o are as defined above, (i) -COR9, wherein R9 is as defined above, (j) -CO2R9, wherein R9 is as defined above, (k) heterocycle, wherein the heterocycle is as defined above;
(4) C2-6alkynyl;
(5) phenyl, unsubstitued or substituted with one or more of the substituent(s) selected from:
(a) hydroxy, (b) Ci-ealkoxy,
(c) Ci-βalkyl,
(d) C2-5alkenyl,
(e) halo,
(f) -CN, (g) -NO2j
(h) -CF3, (i) -(CH2)m-NR9Rio, wherein m, R9 and R are as defined above,
(j) -NR9COR °, wherein R9 and Rio are as defined above,
(k) -NR9CO2Ri°, wherein R9 and Rio are as defined above, (1) -CONR9R10, wherein R9 and Rio are as defined above,
(m) -CO2NR9Ri°, wherein R9 and R10 are as defined above,
(n) -COR9, wherein R9 is as defined above,
(0) -CO2R9, wherein R9 is as defined above;
R2 and R3 are independently selected from the group consisting of:
(1) hydrogen;
(2) Ci-βalkyl, unsubstituted or substituted with one or more of the substituents selected from:
(a) hydroxy, (b) oxo,
(c) Ci-βalkoxy,
(d) phenyl- Ci-3alkoxy,
(e) phenyl,
(f) -CN, (g) halo,
(h) -NR9Ri°, wherein R9 and R10 are independently selected from:
(i) -NR9COR10, wherein R9 and R10 are as defined above,
(j) -NR9CO2Ri°, wherein R9 and Rio are as defined above, (k) -CONR9Ri°, wherein R9 and Rio are as defined above,
(1) -COR9, wherein R9 is as defined above, and (m) -CO2R9, wherein R9 is as defined above;
(3) C2-6alkenyl, unsubstituted or substituted with one or more of the substituent(s) selected from: (a) hydroxy,
(b) oxo, (c) Ci.ealkoxy,
(d) phenyl-Cj..3alkoxy,
(e) phenyl,
(f) -CN, (g) halo,
(h) -CONR9Ri° wherein R9 and R ° are as defined above, (i) -COR9, wherein R9 is as defined above, (j) -CO2R9, wherein R9 is as defined above; (4) C2-ealkynyl; (5) phenyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy,
(b) Ci-ealkoxy,
(c) Ci-ealkyl, (d) C2-5alkenyl,
(e) halo,
(f) -CN, (h) -CFs, (i) -(CH2)m-NR9R °, wherein m, R9 and Rio are as defined above,
(j) -NR9CORi°, wherein R9 and Rio are as defined above,
(k) -NR9CO2Ri°, wherein R9 and Rio are as defined above,
(1) -CONR9Ri°, wherein R9 and Ri are as defined above, (m) -CO2NR9R10, wherein R9 and R10 are as defined above,
(n) -COR9, wherein R9 is as defined above,
(o) -CO2R9, wherein R9 is as defined above;
and the groups R1 and R2 may be joined together to form a heterocyclic ring selected from the group consisting of: (a) pyrrolidinyl, (b) piperidinyl,
(c) pyrrolyl,
(d) pyridinyl,
(e) imidazolyl, (f) oxazolyl, and
(g) thiazolyl, and wherein the heterocyclic ring is unsubstituted or substituted with one or more substituent(s) selected from: (i) Ci-βalkyl, (ii) oxo,
(iii) Ci-βalkoxy,
(iv) -NR9R10, wherein R9 and R10 are as defined above, (v) halo, and (vi) trifluoromethyl;
and the groups R2 and R3 may be joined together to form a carbocyclic ring selected from the group consisting of:
(a) cyclop entyl,
(b) cyclohexyl, (c) phenyl, and wherein the carbocyclic ring is unsubstituted or substituted with one or more substituents selected from:
(i) Ci-βalkyl,
(ii) Ci-ealkoxy, (iii) -NR9Rio, wherein R9 and R10 are as defined above,
(iv) halo, and
(v) trifluoromethyl;
and the groups R2 and R3 may be joined together to form a heterocyclic ring selected from the group consisting of: (a) pyrrolidinyl, (b) piperidinyl,
(c) pyrrolyl,
(d) pyridinyl,
(e) imidazolyl, (f) furanyl,
(g) oxazolyl, (h) thienyl, and (i) thiazolyl, and wherein the heterocyclic ring is unsubstituted or substituted with one or more substituent(s) selected from:
(i) Ci-ealkyl, (ii) oxo, (iii) Ci-calkoxy,
(iv) -NR9R °, wherein R9 and Ri° are as defined above, (v) halo, and
(vi) trifluoromethyl;
X is selected from the group consisting of:
(1) -o-, (2) -S-,
(3) -SO-, and
R4 is selected from the group consisting of: (1)
(2) -Y-Ci-βalkyl, wherein alkyl is unsubstituted or substituted with one or more of the substituents selected from: (a) hydroxy,
(b) oxo,
(c) Ci-βalkoxy,
(d) phenyl-C1-3alkoxy, (e) phenyl,
(f) -CN,
(g) halo,
(h) -NR9R10, wherein R9 and RIO are as defined above, (i) -NR9CORi°, wherein R9 and R10 are as defined above, G) -NR9CO2R10, wherein R9 and R ° are as defined above,
(k) -CONR9Ri°, wherein R9 and Ri° are as defined above, (1) -COR9, wherein R9 is as defined above, (m) -CO2R9, wherein R9 is as defined above;
(3) -Y-C2-6alkenyl, wherein the alkenyl is unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy,
(b) oxo,
(c) d-βalkoxy,
(d) phenyl- d-salkoxy, (e) phenyl,
(f) -CN,
(g) halo,
(h) -CONR9Ri°, wherein R9 and Ri° are as defined above, (i) -COR9, wherein R9 is as defined above, (j) -CO2R9, wherein R9 is as defined above,
(4) -0(CO)-phenyl, wherein the phenyl is unsubstituted or substituted with one or more of R6, R7and R8;
R5 is selected from the group consisting of: (1) phenyl, unsubstituted or substituted with one or more of Ru, Ri2 and R13 ; (2) Ci-salkyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy,
(b) oxo, (c) Cj.-6alkoxy,
(d) phenyl-Cι-3alkoxy,
(e) phenyl,
(f) -CN,
(g) halo, (h) -NR9R °, wherein R9 and Ri° are as defined above,
(i) -NR9CORi°, wherein R9 and Ri° are as defined above, (j) -NR9CO2R10, wherein R9 and Ri° are as defined above, (k) -CONR9R °, wherein R9 and Ri° are as defined above, (1) -COR9, wherein R9 is as defined above, (m) -CO2R9, wherein R9 is as defined above;
(3) C2-6alkenyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy,
(b) oxo, (c) Ci-ealkoxy,
(d) phenyl-Ci.3alkoxy,
(e) phenyl,
(f) -CN,
(g) halo, (h) -CONR9Rio, wherein R9 and Rio are as defined above,
(i) -COR9, wherein R9 is as defined above, (j) -CO2R9, wherein R9 is as defined above;
(4) heterocycle, wherein the heterocycle is as defined above;
RG, R7 and R8 are independently selected from the group consisting of: (1) hydrogen; (2) Ci-βalkyl, unsubstituted or substituted with one or more of the substituents selected from:
(a) hydroxy,
(b) oxo, (c) Ci-ealkoxy,
(d) phenyl-Ci-3alkoxy,
(e) phenyl,
(f) -CN,
(g) halo, (h) -NR9R10, wherein R9 and Ri° are as defined above,
(i) -NR9CORi°, wherein R9 and R ° are as defined above, (j) -NR9CO2R10, wherein R9 and Ri° are as defined above, (k) -CONR9R °, wherein R9 and R ° are as defined above, (1) -COR9, wherein R9 is as defined above, and (m) -CO2R9, wherein R9 is as defined above;
(3) C2-6alkenyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy,
(b) oxo, (c) Ci-ealkoxy,
(d) phenyl-Ci-βalkoxy,
(e) phenyl,
(f) -CN,
(g) halo, (h) -CONR9R10 wherein R9 and Ri° are as defined above,
(i) -COR9 wherein R9 is as defined above, (j) -CO2R9, wherein R9 is as defined above;
(4) C2-ealkynyl;
(5) phenyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy, (b) Ci-calkoxy,
(c) Ci-βalkyl,
(d) C2-5alkenyl,
(e) halo, (f) -CN,
(g) -NO2, (h) -CF3,
(i) -(CH2)m-NR9R o, wherein m, R9 and Ri° are as defined above, (j) -NR9COR10, wherein R9 and Ri° are as defined above,
(k) -NR9CO2R10, wherein R9 and Ri° are as defined above, (1) -CONR9Ri°, wherein R9 and Ri° are as defined above, (m) -CO2NR9Ri°, wherein R9 and Ri° are as defined above, (n) -COR9, wherein R9 is as defined above; (o) -CO2R9, wherein R9 is as defined above;
(6) halo,
(7) -CN,
(8) -CF3, (10) -SR14, wherein Ru is hydrogen or Ci-salkyl,
(11) -SOR14, wherein Ri4 is as defined above,
(12) -SO2R14, wherein R14 is as defined above,
(13) NR9CORi°, wherein R9 and Ri° are as defined above,
(14) CONR9CORi°, wherein R9 and Ri° are as defined above, (15) NR9R10, wherein R9 and R ° are as defined above,
(16) NR9C02Ri°, wherein R9 and R10 are as defined above,
(17) hydroxy,
(18) Ci-ealkoxy,
(19) COR9, wherein R9 is as defined above, (20) CO2R9, wherein R9 is as defined above, Rii, Ri2 and R13 are independently selected from the definitions of R6, R7 and R8, or -OX;
Y is selected from the group consisting of: (1) a single bond,
(2) -O-,
(3) -S-,
(4) -CO-, (6) -CHR15-, and
(7) -CR15Ri°-, wherein R15 and Ri6 are independently selected from the group consisting of:
(a) Ci-ealkyl, unsubstituted or substituted with one or more of the substituents selected from: (i) hydroxy,
(ii) oxo,
(iii) Ci-βalkoxy,
(iv) phenyl-Ci-salkoxy,
(v) phenyl, (vi) -CN,
(vii) halo,
(viii) -NR9Ri°, wherein R9 and Ri° are as defined above,
(ix) -NR9CORi°, wherein R9 and Ri° are as defined above,
(x) -NR9CO2Ri°, wherein R9 and Ri° are as defined above, (xi) -CONR9Rio, wherein R9 and Rio are as defined above,
(xii) -COR9, wherein R9 is as defined above, and
(xiii) -CO2R9, wherein R9 is as defined above;
(b) phenyl, unsubstituted or substituted with one or more of the substituent(s) selected from: (i) hydroxy,
(ii) Ci-βalkoxy, (iii) Ci-βalkyl,
(iv) C2-5alkenyl,
(v) halo,
(vi) -CN,
(vii) -NO2,
( iii) -CF3,
(ix) -(CH2)m-NR9R °, wherein m, R9 and R ° are as defined above,
(x) -NR9COR °, wherein R9 and Rio are as defined above, (xi) -NR9CO2R10, wherein R9 and R ° are as defined above,
(xii) -CONR9Ri°, wherein R9 and Ri° are as defined above, (xiii) -CO2NR9R10, wherein R9 and R10 are as defined above, (xiv) -COR9, wherein R9 is as defined above, and (xv) -CO2R9, wherein R9 is as defined above;
Z is selected from:
(1) hydrogen,
(2) Ci- alkyl, and
(3) hydroxy, with the proviso that if Y is -O-, Z is other than hydroxy, or if Y is -CHR15-, then Z and Ri5 may be joined together to form a double bond.
Particularly preferred compounds of formula (I) are those wherein: R is selected from the group consisting of:
(1) Ci-βalkyl, substituted with one or more of the substituents selected from:
(a) heterocycle, wherein the heterocycle is selected from the group consisting of:
(A) benzimidazolyl,
(B) imidazolyl, (C) isoxazolyl,
(D) isothiazolyl, (E) oxadiazolyl,
(F) pyrazinyl,
(G) pyrazolyl, (H) pyridyl, (I) pyrrolyl,
(J) tetrazolyl, (K) thiadiazolyl, (L) triazolyl, and (M) piperidinyl, and wherein the heterocycle is unsubstituted or substituted with one or more substituent(s) selected from:
(i) Ci-βalkyl, unsubstituted or substituted with halo, -CF3, -OCH3, or phenyl,
(ii) d-ealkoxy, (iii) oxo,
(iv) thioxo, (v) cyano, (vi) -SCH3, (vii) phenyl, (viii) hydroxy,
(ix) trifluoromethyl,
(x) -(CH2)m-NR9R10, wherein m is 0, 1 or 2, and R9 and Rio areindependently selected from:
(I) hydrogen, (II) Ci-ealkyl,
(III) hydroxyCi-ealkyl, and
(IV) phenyl,
(xi) -NR9CORi°, wherein R9 and R10 are as defined above, and (xii) -CONR9R!0, wherein R9 and Rio are as defined above,
R2 and R3 are independently selected from the group consisting of: (1) hydrogen;
(2) Ci-βalkyl
(3) C2-6alkenyl, and
(5) phenyl;
X is - 0-;
R is
R5 is phenyl, unsubstituted or substituted with halo; R6, R7 and R8 are independently selected from the group consisting of: (1) hydrogen,
(2) Ci-ealkyl,
(3) halo, and
(4) -CF3; Y is -O-; and Z is hydrogen or Cι-4alkyl; and pharmaceutically acceptable salts thereof.
Particularly preferred compounds of formula (I) are:
4-(3-(l,2,4-triazolo)methyl)-2(S)-(3,5-bis(trifluoromethyl)benzyloxy)-3(S)- phenyl-morpholine; 4-(3-(l,2,4-triazolo)methyl)-2(S)-(3,5-bis(trifluoromethyl)benzyloxy)-3(R)- phenyl-morpholine;
4-(3-(5-oxo-lH,4H-l,2,4-triazolo)methyl)-2(S)-(3,5- bis(trifluoromethyl)benzyloxy)-3(S)-phenyl-morpholine; and
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)- 4-(3-(5-oxo-lH,4H-l,2,4-triazolo)methyl)morpholine; or a pharmaceutically acceptable salt thereof. Further preferred NK-1 receptor antagonists are those described in International (PCT) Patent Specification No. WO 95/18124, i.e. compounds of formula (II):
or a pharmaceutically acceptable salt or prodrug thereof, wherein
Ri is hydrogen, halogen, Ci-βalkyl, d-ealkoxy, CF3) NO2, CN, SRa, SORa, SO2Ra, CO2Ra, CONRaRb, C2-6alkenyl, C2-6alkynyl or C1.4alkyl substituted by Cι-4alkoxy, where Ra and Rb each independently represent hydrogen or Cj.. alkyl;
R2 is hydrogen, halogen, Ci-ealkyl, Ci-ealkoxy substituted by Ci.4alkoxy or CF3;
R3 is hydrogen, halogen or CF3;
R4 is hydrogen, halogen, Ci-βalkyl, Ci-βalkoxy, CF3, N02, CN, SRa, SORa, SO2Ra, CO2Ra, CONRaRb, or Cι. alkyl substituted by Ci- alkoxy, where Ra and Rb each independently represent hydrogen or Ci- alkyl;
R5 is hydrogen, halogen, Ci-ealkyl, Ci-ealkoxy substituted by Cι- alkoxy or CF3; R6 is a 5-membered or 6-membered heterocyclic ring containing 2 or
3 nitrogen atoms optionally substituted by =O, =S or a Ci- alkyl group, and optionally substituted by a group of the formula ZNR7R8 where Z is Ci-ealkylene or C3-ecycloalkylene;
R7 is hydrogen, Cι- alkyl, C3-7cycloalkyl or C3-7cycloalkylCi-4alkyl, or C2- alkyl substituted by Cι-4alkoxy or hydroxyl; R8 is hydrogen, Cι.4alkyl, C3-7cycloalkyl or C3-7cycloalkylCι- alkyl, or C2-4alkyl substituted by one or two substituents selected from Cι-4alkoxy, hydroxyl or a 4, 5 or 6 membered heteroaliphatic ring containing one or two heteroatoms selected from N, O and S; or R7, R8 and the nitrogen atom to which they are attached form a heteroaliphatic ring of 4 to 7 ring atoms, optionally substituted by a hydroxy group, and optionally containing a double bond, which ring may optionally contain an oxygen or sulphur ring atom, a group S(O) or S(O)2 or a second nitrogen atom which will be part of a NH or NRC moiety where Rc is Ci.4alkyl optionally substituted by hydroxy or Ci-4alkoxy; or R7, R8 and the nitrogen atom to which they are attached form a non-aromatic azabicyclic ring system of 6 to 12 ring atoms; or Z, R7 and the nitrogen atom to which they are attached form a heteroaliphatic ring of 4 to 7 ring atoms which may optionally contain an oxygen ring atom;
R9a and R9b are each independently hydrogen or Ci- alkyl, or R9a and R9b are joined so, together with the carbon atoms to which they are attached, there is formed a C5-7 ring;
X is an alkylene chain of 1 to 4 carbon atoms optionally substituted by oxo; and
Y is a Cι-4alkyl group optionally substituted by a hydroxyl group; with the proviso that if Y is Cι-4alkyl, R6 is susbstituted at least by a group of formula ZNR7R8 as defined above.
Particularly preferred compounds of formula (II) are those of formula (Ila) and pharmaceutically acceptable salts thereof:
(Ha) wherein:
A1 is fluorine or CF3;
A2 is fluorine or CF3;
A3 is fluorine or hydrogen; and X, Y and R6 are as defined in relation to formula (II).
Particularly preferred compounds of formula (II) include: 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(5-(dimethylamino) methyl-l,2,3-triazol-4-yl)methyl-3-(S)-phenylmorpholine; 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(5-(dimethylamino) methyl-l,2,3-triazol-4-yl)methyl-3-(S)-(4-fluorophenyl)morpholine; and pharmaceutically acceptable salts thereof.
Further preferred NK-1 receptor antagonists are those described in European Patent Specification No. WO 95/23798, i.e. compounds of formula (III):
or a pharmaceutically acceptable salt thereof, wherein: R2 and R3 are independently selected from the group consisting of:
(1) hydrogen,
(2) Ci-ealkyl, unsubstituted or substituted with one or more of the substituents selected from: (a) hydroxy,
(b) oxo,
(c) Ci-βalkoxy,
(d) phenyl-Ci-salkoxy,
(e) phenyl, (f) -CN,
(g) halo,
(h) -NR9Ri°, wherein R9 and R10 are independently selected from:
(i) hydrogen, (ii) Ci-βalkyl,
(iii) hydroxy- Ci-βalkyl, and (iv) phenyl, (i) -NR9CORi°, wherein R9 and Rio are as defined above, (j) -NR9CO2Ri°, wherein R9 and Ri° are as defined above, (k) -CONR9Ri°, wherein R9 and Ri are as defined above,
(1) -COR9, wherein R9 is as defined above, and (m) -CO2R9, wherein R9 is as defined above;
(3) C' 2-Galkenyl, unsubstituted or substituted with one or more of the substituent(s) selected from: (a) hydroxy,
(b) oxo,
(c) Ci-ealkoxy,
(d) phenyl-Cι-3alkoxy,
(e) phenyl, (f) -CN,
(g) halo, (h) -CONR9R ° wherein R9 and Rio are as defined above, (i) -COR9 wherein R9 is as defined above, (j) -CO2R9, wherein R9 is as defined above; (4) C2-6alkynyl; (5) phenyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy,
(b) Ci-calkoxy,
(c) Ci-βal yl, (d) C2-5alkenyl,
(e) halo,
(f) -CN,
(g) -NO2, (h) -CF3, (i) -(CH2)m-NR9R1o, wherein m, R9 and Rio are as defined above,
(j) -NR9COR10, wherein R9 and Rio are as defined above,
(k) -NR9CO2R10, wherein R9 and Rio are as defined above,
(1) -CONR9R o, wherein R9 and Rio are as defined above, (m) -CO2NR9Ri°, wherein R9 and Rio are as defined above,
(n) -COR9, wherein R9 is as defined above,
(o) -CO2R9, wherein R9 is as defined above;
and the groups R2 and R3 may be joined together to form a carbocyclic ring selected from the group consisting of:
(a) cyclopentyl,
(b) cyclohexyl,
(c) phenyl, and wherein the carbocyclic ring is unsubstituted or substituted with one or more substituents selected from:
(i) Ci-calkyl, (ii) Ci-βalkoxy,
(iii) -NR9R10, wherein R9 and R10 are as defined above,
(iv) halo, and
(v) trifluoromethyl;
and the groups R2 and R3 may be joined together to form a heterocyclic ring selected from the group consisting of:
(a) pyrrolidinyl,
(b) piperidinyl, (c) pyrrolyl,
(d) pyridinyl,
(e) imidazolyl,
(f) furanyl,
(g) oxazolyl, (h) thienyl, and
(i) thiazolyl, and wherein the heterocyclic ring is unsubstituted or substituted with one or more substituent(s) selected from: (i) Ci-ealkyl, (ii) oxo,
(iii) Ci-βalkoxy,
(iv) -NR9Ri°, wherein R9 and R10 are as defined above, (v) halo, and (vi) trifluoromethyl;
R6, R7 and R8 are independently selected from the group consisting of:
(1) hydrogen;
(2) Ci-βalkyl, unsubstituted or substituted with one or more of the substituents selected from: (a) hydroxy,
(b) oxo, (c) Ci-ealkoxy,
(d) phenyl-CLsalkoxy,
(e) phenyl,
(f) -CN, (g) halo,
(h) -NR9Ri°, wherein R9 and R10 are as defined above, (i) -NR9COR10, wherein R9 and Rio are as defined above, (j) -NR9CO2R10, wherein R9 and R10 are as defined above, (k) -CONR9R10, wherein R9 and Rio are as defined above, (1) -COR9, wherein R9 is as defined above, and
(m) -CO2R9, wherein R9 is as defined above;
(3) C2-6alkenyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy, (b) oxo,
(c) Ci-βalkoxy,
(d) phenyl-CLβalkoxy,
(e) phenyl,
(f) -CN, (g) halo,
(h) -CONR9R10 wherein R9 and Rio are as defined above, (i) -COR9 wherein R9 is as defined above, (j) -CO2R9, wherein R9 is as defined above;
(4) C2-6alkynyl; (5) phenyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(a) hydroxy,
(b) Ci-ealkoxy,
(c) Ci-βalkyl, (d) C2-5alkenyl,
(e) halo, (f) -CN, (h) -CF3,
(i) -(CH2)m-NR9R10, wherein m, R9 and Rio are as defined above,
(j) -NR9COR °, wherein R9 and R o are as defined above, (k) -NR9CO2R10, wherein R9 and Rio are as defined above, (1) -CONR9Ri°, wherein R9 and Rio are as defined above, (m) -CO2NR9Ri°, wherein R9 and R10 are as defined above, (n) -COR9, wherein R9 is as defined above,
(o) -CO2R9, wherein R9 is as defined above;
(6) halo,
(7) -CN,
(8) -CFs,
(10) -SR14, wherein R14 is hydrogen or Ci-salkyl,
(11) -SOR14, wherein R14 is as defined above,
(12) -SO2R14, wherein RX4 is as defined above,
(13) NR9CORi°, wherein R9 and R10 are as defined above, (14) CONR9CORi°, wherein R9 and Rio are as defined above,
(15) NR9R10, wherein R9 and R10 are as defined above,
(16) NR9CO2R10, wherein R9 and Rio are as defined above,
(17) hydroxy,
(18) Ci-ealkoxy, (19) COR9, wherein R9 is as defined above,
(20) CO2R9, wherein R9 is as defined above,
(21) 2-pyridyl,
(22) 3-pyridyl.
(23) 4-pyridyl, (24) 5-tetrazolyl,
(25) 2-oxazolyl, and (26) 2-thiazolyl;
RU, Ri2 and Ri3 are independently selected from the definitions of R6, R7 and R8, or -OX;
A is selected from the group consisting of:
(1) Ci-βalkyl, unsubstituted or substituted with one or more of the substituents selected from:
(a) hydroxy, (h) oxo,
(c) Ci-ealkoxy,
(d) phenyl-C1-3alkoxy,
(e) phenyl,
(f) -CN, (g) halo, wherein halo is fluoro, chloro, bromo or iodo,
(h) -NR9Ri°, wherein R9 and Ri° are as defined above,
(i) -NR9CORi°, wherein R9 and Ri° are as defined above,
(j) -NR9CO2R10, wherein R9 and Ri° are as defined above,
(k) -CONR9Ri°, wherein R9 and Ri° are as defined above, (1) -COR9, wherein R9 is as defined above, and
(m) -CO2R9, wherein R9 is as defined above;
(2) C2-6alkenyl, unsubstituted or substituted with one or more of the substituent(s) selected from: (a) hydroxy,
(b) oxo,
(c) CLβalkoxy,
(d) phenyl-Cι-3alkoxy,
(e) phenyl, (f) -CN,
(g) halo, (h) -CONR9Ri° wherein R9 and Rio are as defined above, (i) -COR9 wherein R9 is as defined above, and (j) -CO2R9, wherein R9 is as defined above; and (3) Cϋ-βalkynyl;
B is a heterocycle, wherein the heterocycle is selected from the group consisting of:
and wherein the heterocycle may be substituted in addition to -X with one or more substituent(s) selected from:
(i) Ci-βalkyl, unsubstituted or substituted with halo, -CF3, -OCH3, or phenyl,
(ii) Ci-ealkoxy, (iii) oxo, (iv) hydroxy, (v) thioxo, (vi) -SR9, wherein R9 is as defined above,
(vii) halo, (viii) cyano, (ix) phenyl, (x) trifluoromethyl, (xi) -(CH2)m-NR9Rio, wherein m is 0, 1 or 2, and R9 and Rio are as defined above, (xii) -NR9COR10, wherein R9 and R10 are as defined above,
(xiii) -CONR9R10, wherein R9 and R10 are as defined above,
(xiv) -CO2R9, wherein R9 is as defined above, and
(xv) -(CH2)m-OR9, wherein m and R9 are as defined above;
p is 0 or 1;
X is selected from:
(a) -PO(OH)O" • M , wherein M+ is a pharmaceutically acceptable monovalent counterion,
(b) -PO(O-)2 ' 2M+,
(c) -PO(O")2 * D2+, wherein D2+ is a pharmaceutically acceptable divalent counterion,
(d) -CH(R )-PO(OH)O" M+, wherein R4 is hydrogen or Cι-3alkyl, (e) -CH(R4)-PO(O-)2 ' 2M+,
(f) -CH(R4)-PO(O")2 D2+,
(h) -CH(R4)-SO3- M+, (i) -CO-CH2CH2-CO2- M+, (j) -CH(CH3)-O-CO-R5, wherein R5 is selected from the group consisting of:
(i) .NHQ M"
O'
(ϋi) O' 'CO2 'M+
(k) hydrogen, with the proviso that if p is 0 and none of Ru, Ri2 or Ri3 are -OX, then X is other than hydrogen;
Y is selected from the group consisting of:
(1) a single bond,
(2) -O-,
(3) -S-,
(4) -CO-, (5) -CH2-,
(6) -CHR15-, and
(7) -CRi5Ri6-, wherein R15 and R1G are independently selected from the group consisting of: (a) Ci-βalkyl, unsubstituted or substituted with one or more of the substituents selected from:
(i) hydroxy,
(ii) oxo,
(iii) d-βalkoxy, (iv) phenyl-Ci-3alkoxy,
(v) phenyl,
(vi) -CN,
(vii) halo,
(viii) -NR9R10, wherein R9 and R10 are as defined above, (ix) -NR9CORi°, wherein R9 and R10 are as defined above,
(x) -NR9CO2R10, wherein R9 and R10 are as defined above,
(xi) -CONR9R10, wherein R9 and Ri° are as defined above,
(xii) -COR9, wherein R9 is as defined above, and
(xiii) -CO2R9, wherein R9 is as defined above; (b) phenyl, unsubstituted or substituted with one or more of the substituent(s) selected from:
(i) hydroxy,
(ii) Ci-ealkoxy,
(iii) Ci-ealkyl, (iv) C2-5alkenyl,
(v) halo,
(vi) -CN,
(ix) -(CH2)m-NR9Rio, wherein m, R9 and Rio are as defined above, (x) -NR9COR10, wherein R9 and R10 are as defined above,
(xi) -NR9CO2Ri°, wherein R9 and R10 are as defined above,
(xii) -CONR9R10, wherein R9 and R10 are as defined above,
(xiii) -Cθ2NR9Ri°, wherein R9 and R10 are as defined above, (xiv) -COR9, wherein R9 is as defined above, and
(xv) -CO2R9, wherein R9 is as defined above;
Z is selected from:
(1) hydrogen, (2) Ci-βalkyl, and
(3) hydroxy, with the proviso that if Y is -O-, Z is other than hydroxy, or if Y is -CHR15-, then Z and Ri5 may be joined together to form a double bond.
Particularly preferred compounds of formula (III) are those wherein: R2 and R3 are independently selected from the group consisting of:
(1) hydrogen,
(2) Ci-βalkyl.
(3) C2-6alkenyl, and
(4) phenyl; R6, R7 and R8 are independently selected from the group consisting of:
(1) hydrogen,
(2) Ci-ealkyl,
(3) fluoro,
(4) chloro, (5) bromo,
(6) iodo, and
(7) -CF3;
Ru, Ri2 and RX3 are independently selected from the group consisting of: (1) fluoro, (2) chloro,
(3) bromo, and (4) iodo; A is unsubstituted l-ealkyl; B is selected from the group consisting of:
X H X
N-N N-N N-N
X
N ^ > x
X H N
p is 0 or 1;
X is selected from:
(a) -PO(OH)O" • M+, wherein M+ is a pharmaceutically acceptable monovalent counterion, (b) -PO(O")2 2M+,
(c) -PO(0")2 * D2+, wherein D2+ is a pharmaceutically acceptable divalent counterion,
(d) -CH(R4)-PO(OH)0" • M+, wherein R4 is hydrogen or Cι-3alkyl. (e) -CH(R4)-PO(O-)2 2M+,
(f) -CH(R4)-PO(O-)2 D +, (i) -CO-CH2CH2-CO2- M+,
(j) -CH(CH3)-O-CO-R5, wherein R5 is selected from the group consisting of:
(i) . NH3 M"
O
H2 M"
(ϋ) -N?
O' OH
(ϋi) 0 C02 " M
C02 " M+
(iv)
0 C02 " M+
COQ
(v) "O
NHQ
Y is -O-;
Z is hydrogen or Ci-calkyl; and pharmaceutically acceptable salts thereof. Particularly preferred compounds of formula (III) include:
(1) 2-(S)-(3,5-bis(trifluoromethyl)benzyloxy)-3-(S)-phenyl-4-(3-(5-oxo- lH,4H-l,2,4-triazolo)methyl)morpholine N-oxide;
(2) 2-(S)-(3,5-bis(trifluoromethyl)benzyloxy)-3-(S)-phenyl-4-(3-(4- (ethoxycarbonyloxy-l-ethyl)-5-oxo-lH-l,2,4- tr iazolo) methyl) morp holine ;
(3) 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4- fluorophenyl)-4-(3-(4-monophosphoryl-5-oxo-lH- 1,2,4- triazolo)methyl)morpholine; (4) 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4- fluorophenyl)-4-(3-(l-monophosphoryl-5-oxo-lH- 1,2,4- tr iazolo)methy 1) morp holine ;
(5) 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4- fluorophenyl)-4-(3-(2-monophosphoryl-5-oxo-lH- 1,2,4- triazolo)methyl)morpholine;
(6) 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4- fluorophenyl)-4-(3-(5-oxyphosphoryl-lH-l,2,4- triazolo)methy 1) morp holine ;
(7) 2-(S)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4- fluorophenyl)-4-(3-(l-monophosphoryl-5-oxo-4H- 1,2,4- triazolo) methyl) morp holine ; and pharmaceutically acceptable salts thereof.
Further preferred NK-1 receptor antagonists are those described in European Patent Specification No. WO 96/05181, i.e. compounds of formula (IV):
wherein
X is a group of the formula ΝR6R7 or a C- or N-linked imidazolyl ring;
Y is hydrogen or C1.4alkyl optionally substituted by a hydroxy group;
R is hydrogen, halogen, d-ealkyl, d-ealkoxy, CF3, NO2, CN, SRa, SORa, SO2Ra, CO2Ra, CONRaRb, C^alkenyl, C2-Galkynyl or d- alkyl substituted by d- alkoxy, wherein Ra and Rb each independently represent hydrogen or Cι- alkyl;
R2 is hydrogen, halogen, d-βalkyl, Ci-ealkoxy substituted by Ci- alkoxy or CF3;
R3 is hydrogen, halogen or CF3; R4 is hydrogen, halogen, d-ealkyl, C1.6alkoxy, hydroxy, CF3, NO2,
CN, SRa, SORa, SO2Ra, CO2Ra, CONRaR , C2-6alkenyl, C2-6alkynyl or Cι- alkyl substituted by C1. alkoxy, wherein Ra and Rb are as previously defined;
R5 is hydrogen, halogen, Ci-Galkyl, d-6alkoxy substituted by Cι_4alkoxy or CF3;
RG is hydrogen, d-Galkyl, C3-7cycloalkyl, C3-7cycloalkylCι- alkyl, phenyl, or C2-4alkyl substituted by Ci-4alkoxy or hydroxy;
R7 is hydrogen, d-ealkyl, C3.7cycloalkyl, C3-7cycloalkylCι-4alkyl, phenyl, or C2. alkyl substituted by one or two substituents selected from Ci-4alkoxy, hydroxy or a 4, 5 or 6 membered heteroaliphatic ring containing one or two heteroatoms selected from N, O and S; or R6 and R7, together with the nitrogen atom to which they are attached, form a saturated or partially saturated heterocyclic ring of 4 to 7 ring atoms, which ring may optionally contain in the ring one oxygen or sulphur atom or a group selected from NR8, S(O) or S(O)2 and which ring may be optionally substituted by one or two groups selected from hydroxyC1.4alkyl, C1. alkoxyCι- alkyl, oxo, CORa or CO2Ra where Ra is as previously defined; or R6 and R7 together with the nitrogen atom to which they are attached, form a non-aromatic azabicyclic ring system of 6 to 12 ring atoms;
R8 is hydrogen, d-4alkyl, hydroxy Ci- alkyl or d-4alkoxyd.4alkyl; and R9a and R9b are each independently hydrogen or C1. alkyl, or R9a and
R9b are joined so, together with the carbon atoms to which they are attached, there is formed a C5-7 ring; and pharmaceutically acceptable salts thereof.
Particularly preferred compounds of formula (IV) are those of formula (IVa) and pharmaceutically acceptable salts thereof:
wherein
A1 is fluorine or CF3; A2 is fluorine or CF3;
A3 is fluorine or hydrogen; and X and Y are as defined in relation to formula (I).
Specific compounds of formula (IV) of use in the present invention include:
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-
4-(4-morpholinobut-2-yn-yl)morpholine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(4-N,N- dimethylaminobut-2-yn-yl)-3-(S)-(4-fluorophenyl)morpholine; 4-(4-azetidinylbut-2-yn-yl)-2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl) ethoxy)-3-(S)-(4-fluorophenyl)morpholine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-
4-(4-imidazolylbut-2-yn-yl)morpholine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)- 4-(4-(N-methylpiperazinyl)but-2-yn-yl)morpholine;
4-(4-bis(2-methoxyethyl)aminobut-2-yn-yl)-2-(R)-(l-(R)-(3,5- bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)morpholine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-
4-(4-pyrrolidinobut-2-yn-yl)morpholine; 3-(S)-(4-fluorophenyl)-2-(R)-(l-(R)-(3-fluoro-5-(trifluoromethyl)phenyl) ethoxy)-4-(4-morpholinobut-2-yn-yl)morp holine;
3-(S)-(4-fluorophenyl)-4-(4-morpholinobut-2-yn-yl)-2-(R)-(l-(R)-(3-
(trifluoromethyl)phenyl)ethoxy)morpholine;
4-(4-azetidinylbut-2-yn-yl)-3-(S)-(4-fluorophenyl)-2-(R)-(l-(R)-(3- (trifluoromethyl)phenyl)ethoxy)morpholine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(4-(N-(2- methoxyethyl)-N-methyl)aminobut-2-yn-yl)-3-(S)-phenylmorpholine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(4-(N-cyclopropyl-N-
(2-methoxyethyl)amino)but-2-yn-yl)-3-(S)-phenylmorpholine; 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(4-(N-isopropyl-N-(2- methoxyethyl)amino)but-2-yn-yl)-3-(S)-phenylmorpholine; 4-(4-(N,N-dimethylamino)but-2-yn-yl)-3-(S)-(4-fluorophenyl)-2-(R)-(l-(S)-(3- fluoro-5-(trifluoromethyl)phenyl-2-hydroxyethoxy)morpholine;
4-(4-azetidinylbut-2yn-yl)-3-(S)-(4-fluorophenyl)-2-(R)-(l-(S)-(3-fluoro-5-
(trifluoromethyl)phenyl)-2-hydroxyethoxy)morpholine; 2-(R)-(l-(S)-(3,5-bis(trifluoromethyl)phenyl)-2-hydroxyethoxy)-4-(4-(N,N- dimethylamino)but-2-yn-yl)-3-(S)-(4-fluorophenyl)morp holine;
4-(4-azetidinylbut-2-yn-yl)-2-(R)-(l-(S)-(3,5-bis(trifluoromethyl)phenyl-2- hydroxyethoxy)-3-(S)-(4-fluorophenyl)morpholine;
4-(4-N-bis(2-methoxy)ethyl-N-methylamino)but-2-yn-yl)-2-(R)-(l-(R)-(3,5- bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)morpholine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-
4-(4-(2-(S)-(methoxymethyl)pyrrolidino)but-2-yn-yl)morpholine;
4-(4-(7-azabicyclo[2.2.1]heptano)but-2-yn-yl)-2-(R)-(l-(R)-(3,5- bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)morpholine; 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(4- diisopropylaminobut-2-yn-yl)-3-(S)-(4-fluorophenyl)morpholine;
2-(R)-(l-(R)-(3-fluoro-5-(trifluoromethyl)phenyl)ethoxy)-4-(4-(2-(S)-
(methoxymethyl)pyrrolidino)but-2-yn-yl)-3-(S)-phenylmorphohne;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)- 4- (4- (2- (S) - (hy droxymethyl)pyrrolidino)but-2-y n-y l)morpholine ; and pharmaceutically acceptable salts thereof.
Another class of NK-1 receptor antagonists of use in the present invention is that described in European Patent Specification No.
0 436 334, i.e. compounds of formula (V):
or a pharmaceutically acceptable salt thereof, wherein
Y is (CH2)n wherein n is an integer from 1 to 4, and wherein any one of the carbon-carbon single bonds in said (CH2)n may optionally be replaced by a carbon-carbon double bond, and wherein any one of the carbon atoms of said (CH2)n may optionally be substituted with R4, and wherein any one of the carbon atoms of said (CH2)n may optionally be substituted with R7;
Z is (CH2)m wherein m is an integer from 0 to 6, and wherein any one of the carbon-carbon single bonds of (CH2) may optionally be replaced by a carbon-carbon double bond or a carbon-carbon triple bond, and any one of the carbon atoms of said (CH2)m may optionally be substituted with R8;
Ri is hydrogen or Ci-salkyl optionally substituted with hydroxy, Cι-4alkoxy or fluoro; R2 is a radical selected from hydrogen, d-e straight or branched alkyl, C3-7cycloalkyl wherein one of the CH2 groups in said cycloalkyl may optionally be replaced by NH, oxygen or sulphur; aryl selected from phenyl and naphthyl; heteroaryl selected from indanyl, thienyl, furyl, pyridyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl and quinolyl; phenyl- C2-6alkyl, benzhydryl and benzyl, wherein each of said aryl and heteroaryl groups and the phenyl moieties of said benzyl, phenyl - C2-6 lkyl and benzhydryl may optionally be substituted with one or more substituents independently selected from halo, nitro, d-β alkyl, Ci-ealkoxy, trifluoromethyl, amino, Cι-6alkylamino, Ci-βalkyl-O-CO, Ci-ealkyl-O-CO- d-ealkyl, d-ealkyl-CO-O, d-ealkyl-CO-d-ealkyl-O-, Ci-ealkyl-CO, d-ealkyl-CO-Ci-ealkyl-, di-Ci.ealkylamino, -CONH-Ci-ealkyl, Ci-ealkyl-CO-NH-Ci-ealkyl, -NHCOH and -NHCO-Ci-ealkyl; and wherein one of the phenyl moieties of said benzhydryl may optionally be replaced by naphthyl, thienyl, furyl or pyridyl; R5 is hydrogen, phenyl or Ci.ealkyl; or R2 and R5 together with the carbon to which they are attached, form a saturated ring having from 3 to 7 carbon atoms wherein one of the CH2 groups in said ring may optionally be replaced by oxygen, NH or sulfur; R3 is aryl selected from phenyl and naphthyl; heteroaryl selected from indanyl, thienyl, furyl, pyridyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl and quinolyl; and cycloalkyl having 3 to 7 carbon atoms wherein one of the (CH2) groups in said cycloalkyl may optionally be replaced by NH, oxygen or sulphur; wherein each of said aryl and heteroaryl groups may optionally be substituted with one or more substituents, and said C3-7cycloalkyl may optionally be substituted with one or two substituents, each of said substituents being independently selected from halo, nitro, Ci-calkyl, Ci-ealkoxy, trifluoromethyl, amino, Ci-ealkylamino, -CO-NH- Ci-ealkyl, Ci-ealkyl-CO-NH-Ci-ealkyl, -NHCOH and -NHCO-Ci-ealkyl;
R4 and R7 are each independently selected from hydroxy, halogen, halo, amino, oxo, cyano, methylene, hydroxymethyl, halomethyl, Ci-ealkylamino, di-d-ealkylamino, Ci-ealkoxy, Cι-6alkyl-O-CO, Ci-ealkyl-O-CO-d-ealkyl, Ci-ealkyl-CO-O, Ci-ealkyl-CO-Ci-ealkyl-O-, Ci-ealkyl-CO-, Ci-Galkyl-CO-d-ealkyl, and the radicals set forth in the definition of R2;
Ro is -NHCOR9, -NHCH2R9, SO2R8 or one of the radicals set forth in any of the definitions of R2, R4 and R7;
R8 is oximino (=NOH) or one of the radicals set forth in any of the definitions of R2, R4 and R7;
R9 is Ci-ealkyl, hydrogen, phenyl or phenylCι-6alkyl; with the proviso that (a) when m is 0, R8 is absent, (b) when R4, RG, R7 or R8 is as defined in R2, it cannot form together with the carbon to which it is attached ,a ring with R5, and (c) when R4 and R7 are attached to the same carbon atom, then either each of R4 and R7 is independently selected from hydrogen, fluoro and Ci-ealkyl, or R4 and R7, together with the carbon to which they are attached, for a C3-6 saturated carbocyclic ring that forms a spiro compound with the nitrogen-containing ring to which they are attached.
A particularly preferred compound of formula (V) is (2S,3S)-cis-3-(2- methoxybenzylamino)-2-phenylpiperidine; or a pharmaceutically acceptable salt thereof.
Another class of NK-1 receptor antagonists of use in the present invention is that described in International Patent Specification No. WO 93/21155, i.e. compounds of formula (VI):
or a pharmaceutically acceptable salt thereof, wherein radicals R are phenyl radicals optionally 2- or 3-substituted by a halogen atom or a methyl radical; Ri is optionally substituted phenyl, cyclohexadienyl, naphthyl, indenyl or optionally substituted heterocycle;
R2 is H, halogen, OH, alkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, alkyloxy, alkylthio, acyloxy, carboxy, optionally substituted alkyloxycarbonyl, benzyloxycarbonyl, amino or acylamino; R3 is optionally 2-substituted phenyl;
R4 is OH or fluorine when R5 is H; or R4 and R5 are OH ; or R4 and R5 together form a bond.
A particularly preferred compound of formula (VI) is (3aS, 4S, 7aS)- 7,7-diphenyl-4-(2-methoxyphenyl)-2-[(2S)-(2-methoxyphenyl)propionyl] perhydroisoindol-4-ol; or a pharmaceutically acceptable salt thereof. Another class of NK-1 receptor antagonists of use in the present invention is that described in European Patent Specification No. 0 591 040, i.e. compounds of formula (VII):
R Q
Ar-T- CO-N-CH2 - C-CH2 -CH2 -Am + , A " (VII)
Ar ' wherein
Ar represents an optionally substituted mono-, di- or tricyclic aromatic or heteroaromatic group;
T represents a bond, a hydroxymethylene group, a Cι-4alkoxymethylene group or a Ci-salkylene group; Ar' represents a phenyl group which is unsubstituted or substituted by one or more substituents selected from halogen, preferably chlorine or fluorine, trifluoromethyl, d-4alkoxy, Cι-4alkyl where the said substituents may be the same or different; a thienyl group; a benzothienyl group; a naphthyl group; or an indolyl group; R represents hydrogen, Cι. alkyl, ω-Cι. alkoxyCι- alkyl, or ω - C2-4alkanoyloxy C2-4alkyl;
Q represents hydrogen; or Q and R together form a 1,2-ethylene, 1,3-propylene or 1,4- butylene group; Am+ represents the radical
in which Xi, X2 and X3, together with the nitrogen atom to which they are attached, form an azabicyclic or azatricyclic ring system optionally substituted by a phenyl or benzyl group; and A- represents a pharmaceutically acceptable anion. A particularly preferred compound of formula (VII) is (+) l-[2-[3- (3,4-dichlorophenyl)-l-[(3-isopropoxyphenyl)acetyl]-3-piperidinyl]ethyl]-4- phenyl-l-azabicyclo[2,2,2]octane; or a pharmaceutically acceptable salt, especially the chloride, thereof. Another class of NK-1 receptor antagonists of use in the present invention is that described in European Patent Specification No. 0 532 456, i.e. compounds of formula (VIII):
or a pharmaceutically acceptable salt thereof, wherein Ri represents an optionally substituted aralkyl, aryloxyalykl, heteroaralkyl, aroyl, heteroaroyl, cycloalkylcarbonyl, aralkanoyl, heteroarylalkanoyl, aralkoxycarbonyl or arylcarbamoyl group or the acyl group of an α-amino acid optionally N-substituted by a lower alkanoyl or carbamoyl-lower alkanoyl group; R2 represents cycloalkyl or an optionally substituted aryl or heteroaryl group;
R3 represents hydrogen, alkyl, carbamoyl or an alkanoyl or alkenoyl group optionally substituted by carboxy or esterified or amidated carboxy;
R4 represents an optionally substituted aryl group or an optionally partially saturated heteroaryl group;
Xi represents methylene, ethylene, a bond, an optionally ketalised carbonyl group or an optionally etherified hydroxymethylene group;
X2 represents alkylene, carbonyl or a bond; and
X3 represents carbonyl, oxo-lower alkyl, oxo(aza)-lower alkyl, or an alkyl group optionally substituted by phenyl, hydroxymethyl, optionally esterified or amidated carboxy, or (in other than the α-position) hydroxy.
A particularly preferred compound of formula (VIII) is (2R*, 4S*)-2- benzyl-l-(3,5-dimethylbenzoyl)-N-(4-quinolinylmethyl)-4-piperidineamine; or a pharmaceutically acceptable salt thereof. Another class of NK-1 receptor antagonists of use in the present invention is that described in European Patent Specification No. 0 443 132, i.e. compounds of formula (IX)
or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, or a group of the formula:
X is CH or N; and Z is O or N-R5, in which R5 is hydrogen or lower alkyl;
R2 is hydroxy or lower alkoxy;
R3 is hydrogen or optionally substituted lower alkyl;
R4 is optionally substituted ar(lower)alkyl;
A is carbonyl or sulfonyl; and Y is a bond or lower alkenylene.
A particularly preferred compound of formula (IX) is the compound of formula (IXa)
HO
or a pharmaceutically acceptable salt thereof.
Another class of NK-1 receptor antagonists of use in the present invention is that described in International Patent Specification No. WO 92/17449, i.e. compounds of the formula (X)
or a pharmaceutically acceptable salt thereof, wherein
R1 is aryl selected from indanyl, phenyl and naphthyl; heteroaryl selected from thienyl, furyl, pyridyl and quinolyl; and cycloalkyl having 3 to 7 carbon atoms, wherein one of said carbon atoms may optionally be replaced by nitrogen, oxygen or sulfur; wherein each of said aryl and heteroaryl groups may optionally be substituted with one or more substituents, and said C3-7cycloalkyl may optionally be substituted with one or two substituents, said substituents being independently selected from chloro, fluoro, bromo, iodo, nitro, Ci-ioalkyl optionally substituted with from one to three fluoro groups, Ci-ioalkoxy optionally substituted with from one to three fluoro groups, amino, Ci-ioalkyl-S-, Cι-!oalkyl-S(O)-, Cι-ιoalkyl-SO2-, phenyl, phenoxy, Cι-ιoalkyl-SO2NH-,
Cι-ιealkyl-Sθ2NH-Cι-ι0akyl-, d-ιoalkylamino-diCι-ιoalkyl-, cyano, hydroxy, cycloalkoxy having 3 to 7 carbon atoms, Ci-ealkylamino, Ci-edialkylamino, HC(O)NH- and Cι-ιoalkyl-C(0)NH-; and
R2 is thienyl, benzhydryl, naphthyl or phenyl optionally substituted with from one to three substituents independently selected from chloro, bromo, fluoro, iodo, cycloalkoxy having 3 to 7 carbon atoms, Ci-ioalkyl optionally substituted with from one to three fluoro groups and Ci-ioalkoxy optionally substituted with from one to three fluoro groups.
A particularly preferred compound of formula (X) is (2S,3S)-3-(2- methoxy-5-trifluoromethoxybenzyl)-amino-2-phenylpiperidine; or a pharmaceutically acceptable salt thereof. Another class of NK-1 receptor antagonists of use in the present invention is that described in International Patent Specification No. WO 95/08549, i.e. compounds of formula (XI) '
or a pharmaceutically acceptable salt thereof, wherein Ri is a Cι. alkoxy group; R2 is
R3 is a hydrogen or halogen atom; R4 and R5 may each independently represent a hydrogen or halogen atom, or a Cι-4alkyl, Cι-4alkoxy or trifluoromethyl group;
R6 is a hydrogen atom, a Cι-4alkyl, (CH2)mcyclopropyl, -S(0)nCι- 4alkyl, phenyl, NR7R8, CH2C(O)CF3 or trifluoromethyl group;
R7 and R8 may each independently represent a hydrogen atom, or a Cι-4alkyl or acyl group; x represents zero or 1; n represents zero, 1 or 2; and m represents zero or 1.
Particularly preferred compounds of formula (XI) are (2-methoxy-5- tetrazol-l-yl-benzyl)-([2<S,3S]-2-phenyl-piperidin-3-yl)-amine; and [2- methoxy-5-(5-trifluoromethyl-tetrazol-l-yl)-benz3 ]-([2S,3S,]-2-phenyl- piperidin-3-yl)-amine; or a pharmaceutically acceptable salt thereof. Another class of tachykinin antagonists of use in the present invention is that described in International Patent Specification No. WO 95/14017, i.e. compounds of formula (XII)
R8 R4
R— (CH2)n — C- CH2 N- (CE ) — R3 NH R2
(CO)p
I
or a pharmaceutically acceptable salt thereof, wherein m is zero, 1, 2 or 3; n is zero or 1; o is zero, 1 or 2; p is zero or 1; R is phenyl, 2- or 3-indolyl, 2- or 3-indolinyl, benzothienyl, benzofuranyl, or naphthyl; which R groups may be substituted with one or two halo, Cι-3alkoxy, trifluoromethyl, Cι-4alkyl, phenyl-Cι-3alkoxy, or Cι-4alkanoyl groups; Ri is trityl, phenyl, diphenylmethyl, phenoxy, phenylthio, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, indolinyl, indolyl, benzothienyl, hexamethyleneiminyl, benzofuranyl, tetrahydropyridinyl, quinolinyl, isoquinolinyl, reduced quinolinyl, reduced isoquinolinyl, phenyl-(Cι- alkyl)-, phenyl-(d- alkoxy)-, quinolinyl-(Cι-4alkyl)-, isoquinolinyl-(d- alkyl)-, reduced quniolinyl-(Cι- alkyl)-, reduced isoquinolinyl-(Ci- alkyl)-, benzoyl-(Cι-3alkyl)-, Cι.4alkyl, or -NH-CH2-R5; any one of which Rx groups may be substituted with halo, Cι-4alkyl, Cι-4alkoxy, trifluoromethyl, amino, Cι-4alkylamino, di(Cι-4alkyl)amino, or C2-4alkanoylamino ; or any one of which R1 groups may be substituted with phenyl, piperazinyl, C3-8cycloalkyl, benzyl, Cι-4alkyl, piperidinyl, pyridinyl, pyrimidinyl, C2-Galkanoylamino, pyrrolidinyl, d-ealkanoyl, or Cι-4alkoxycarbonyl; any one of which groups may be substituted with halo, d. alkyl, d-4alkoxy, trifluoromethyl, amino, C1.4alkylamino, di(Cι- alkyl)amino, or C2-4alkanoylamino; or R1 is amino, a leaving group, hydrogen, Cι-4alkylamino, or di(C !-4alkyl) amino ;
R5 is pyridyl, anilino-(Cι-3alkyl)-, or anilinocarbonyl;
R2 is hydrogen, d-4alkyl, Cι-4alkylsulfonyl, carboxy-(Cι-3alkyl)-, Ci-salkoxycarbonyMCi-salkyi)-, or -CO-R6;
R6 is hydrogen, Cι-4alkyl, Cι-3haloalkyl, phenyl, Cι-3alkoxy, Cι-3hydroxyalkyl, amino, Cι-4alkylamino, di(Cι-4alkyl)amino, or -(CH2)q-R7; q is zero to 3;
R7 is carboxy, Cι-4alkoxycarbonyl, Cι.4alkylcarbonyloxy, amino, Cι- alkylamino, di(Cι- alkyl)amino, Ci-ealkoxycarbonylamino, or phenoxy, phenylthio, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, indolinyl, indolyl, benzothienyl, benzofuranyl, quinolinyl, phenyl-(Cι-4alkyl)-, quinolinyl- (Cι. alkyl)-, isoquinolinyl-(d-4alkyl)-, reduced quinolinyl- (Cι-4alkyl)-, reduced isoquinolinyl-(Cι.4alkyl)-, benzoyl-Cι-3alkyl; any one of which aryl or heterocyclic R7 groups may be substituted with halo, trifluoromethyl, Cι-4alkoxy, Cι-4alkyl, amino, Cι-4alkylamino, di(Ci.4alkyl) amino, or C2-4alkanoylamino; or any one of which R7 groups may be substituted with phenyl, piperazinyl, C3-8cycloalkyl, benzyl, piperidinyl, pyridinyl, pyrimidinyl, pyrrolidinyl, d ealkanoyl, or Cι-4alkoxycarbonyl; any of which groups may be substituted with halo, trifluoromethyl, amino, d-4alkoxy, Cι-4alkyl, d-4alkylamino, di(Cι-4alkyl)amino, or C2. alkanoylamino ;
R8 is hydrogen or Ci-ealkyl; R3 is phenyl, phenyl-(Cι-6alkyl)-, C3-8cycloalkyl, Cs-scycloalkenyl,
Ci-βalkyl, naphthyl, C2-8alkenyl, or hydrogen; any one or which groups except hydrogen may be substituted with one or two halo, d-3alkoxy, Cι-3alkylthio, nitro, trifluoromethyl, or Cι-3alkyl groups; and
R4 is hydrogen or Cι-3alkyl; with the proviso that if Ri is hydrogen or halo, R3 is phenyl, phenyl-(d-6alkyl)-, C3-8cycloalkyl, Cs-scycloalkenyl, or naphthyl.
A particularly preferred compound of formula (XII) is [N-(2- methoxybenzyl)acetylamino]-3-(lH-indol-3-yl)-2-[N-(2-(4-piperidin-l- yl)piperidin-l-yl)acetylamino]propane; or a pharmaceutically acceptable salt thereof.
The preferred compounds of formulae (I), (II), (III) and (IV) will have the 2- and 3-substituents on the morpholine ring in the cis arrangement, the preferred stereochemistry being as shown in the following general formula:
Where the benzyloxy moiety is α-substituted, the preferred stereochemistry of the α-carbon is either (R) when the substituent is an alkyl (e.g. methyl) group or (S) when the substituent is a hydroxyalkyl (e.g. hydroxymethyl) group.
Unless otherwise defined herein, suitable alkyl groups include straight-chained and branched alkyl groups containing from 1 to 6 carbon atoms. Typical examples include methyl and ethyl groups, and straight - chained or branched propyl and butyl groups. Particular alkyl groups are methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl and tert-butyl. Unless otherwise defined herein, suitable alkenyl groups include straight-chained and branched alkenyl groups containing from 2 to 6 carbon atoms. Typical examples include vinyl and allyl groups.
Unless otherwise defined herein, suitable alkynyl groups include straight-chained and branched alkynyl groups containing from 2 to 6 - carbon atoms. Typical examples include ethynyl and propargyl groups.
Unless otherwise defined herein, suitable cycloalkyl groups include groups containing from 3 to 7 carbon atoms. Particular cycloalkyl groups are cyclopropyl and cyclohexyl. Unless otherwise defined herein, suitable aryl groups include phenyl and naphthyl groups.
A particular aryl-C1 6alkyl, e.g. phenyl-Ci-ealkyl, group is benzyl.
Unless otherwise defined herein, suitable heteroaryl groups include pyridyl, quinolyl, isoquinolyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyranyl, furyl, benzofuryl, thienyl, benzthienyl, imidazolyl, oxadiazolyl and thiadiazolyl groups.
The term "halogen" as used herein includes fluorine, chlorine, bromine and iodine.
The compounds of use in this invention may have one or more asymmetric centres and can therefore exist as enantiomers and possibly as diastereoisomers. It is to be understood that the present invention relates to the use of all such isomers and mixtures thereof.
Suitable pharmaceutically acceptable salts of the NK-1 receptor antagonists of use in the present invention include acid addition salts which may, for example, be formed by mixing a solution of the compound with a solution of a pharmaceutically acceptable non-toxic acid such as hydrochloric acid, fumaric acid, maleic acid, succinic acid, acetic acid, citric acid, tartaric acid, carbonic acid, phosphoric acid or sulphuric acid. Salts of amine groups may also comprise the quaternary ammonium salts in which the amino nitrogen atom carries an alkyl, alkenyl, alkynyl or aralkyl group. Where the compound carries an acidic group, for example a carboxylic acid group, the present invention also contemplates salts thereof, preferably non-toxic pharmaceutically acceptable salts thereof, such as the sodium, potassium and calcium salts thereof.
Suitable pharmaceutically acceptable salts of the anorectic agent of use in the combinations of the present invention include those salts described above in relation to the salts of NK-1 receptor antagonists. The present invention accordingly provides the use of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII), for the manufacture of a medicament for the treatment or prevention of eating disorders.
The present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an effective amount of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII).
In a further aspect of the present invention, there is provided a pharmaceutical composition for the treatment or prevention of eating disorders comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII), together with at least one pharmaceutically acceptable carrier or excipient.
It will be appreciated that the NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) may be used in combination with an anorectic agent, present as a combined preparation for simultaneous, separate or sequential use for the treatment or prevention of eating disorders. Such combined preparations may be, for example, in the form of a twin pack.
In a further or alternative aspect of the present invention, there is therefore provided a product comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
In a preferred aspect, the present invention accordingly provides the use of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a - halogenated amphetamine derivative for the manufacture of a medicament for the treatment or prevention of eating disorders.
The present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative such that together they give effective relief.
In a further aspect of the present invention, there is provided a pharmaceutical composition comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative together with at least one pharmaceutically acceptable carrier or excipient. In a further or alternative aspect of the present invention, there is provided a product comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
Particularly preferred halogenated amphetamine derivatives are selected from the group consisting of fenfluramine and dexfenfluramine. Thus in a further preferred aspect, the present invention accordingly provides the use of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, for the manufacture of a medicament for the treatment or prevention of eating disorders.
The present invention also provides a method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment an amount of a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and an amount of a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, such that together they give effective relief.
In a further aspect of the present invention, there is provided a pharmaceutical composition comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, together with at least one pharmaceutically acceptable carrier or excipient.
In a further or alternative aspect of the present invention, there is provided a product comprising a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) and a halogenated amphetamine derivative selected from the group consisting of fenfluramine and dexfenfluramine, as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
As stated above, the NK-1 receptor antagonist and the anorectic agent may be formulated in a single pharmaceutical composition or alternatively in individual pharmaceutical compositions for simultaneous, separate or sequential use in accordance with the present invention. Preferably the compositions according to the present invention are in unit dosage forms such as tablets, pills, capsules, powders, granules, solutions or suspensions, or suppositories, for oral, parenteral or rectal administration, by inhalation or insufflation or administration by trans - dermal patches or by buccal cavity absorption wafers. Oral dosage forms are particularly preferred (e.g. tablets, capsules, pills or wafers). For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical carrier, e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a non-toxic pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention. The tablets or pills of the novel composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
The liquid forms in which the novel compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavoured syrups, aqueous or oil suspensions, and flavoured emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, peanut oil or soybean oil, as well as elixirs and similar pharmaceutical vehicles. Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone or gelatin.
Preferred compositions for administration by injection include those comprising a NK-1 receptor antagonist as the active ingredient, in association with a surface-active agent (or wetting agent or surfactant) or in the form of an emulsion (as a water-in-oil or oil-in-water emulsion). Suitable surface-active agents include, in particular, non-ionic agents, such as polyoxyethylenesorbitans (e.g. Tween™ 20, 40, 60, 80 or 85) and other sorbitans (e.g. Span™ 20, 40, 60, 80 or 85). Compositions with a surface -active agent will conveniently comprise between 0.05 and 5% surface-active agent, and preferably between 0.1 and 2.5%. It will be appreciated that other ingredients may be added, for example mannitol or other pharmaceutically acceptable vehicles, if necessary.
Suitable emulsions may be prepared using commercially available fat emulsions, such as Intralipid™, Liposyn™, Infonutrol™, ipofundin™ and Lipiphysan™. The active ingredient may be either dissolved in a pre- mixed emulsion composition or alternatively it may be dissolved in an oil (e.g. soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e.g. egg phospholipids, soybean phospholipids or soybean lecithin) and water. It will be appreciated that other ingredients may be added, for example glycerol or glucose, to adjust the tonicity of the emulsion. Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%. The fat emulsion will preferably comprise fat droplets between 0.1 and l.Oμm, particularly 0.1 and 0.5μm, and have a pH in the range of 5.5 to 8.0. Particularly preferred emulsion compositions are those prepared by mixing a NK-1 receptor antagonist selected from the compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) with Intralipid™ or the components thereof (soybean oil, egg phospholipids, glycerol and water).
Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above. Preferably the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in preferably sterile pharmaceutically acceptable solvents may be nebulised by use of inert gases. Nebulised solutions may be breathed directly from the nebulising device or the nebulising device may be attached to a face mask, tent or intermittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
Compositions of the present invention may also be presented for administration in the form of trans-dermal patches using conventional technology. The compositions may also be administered via the buccal cavity using, for example, absorption wafers.
The present invention further provides a process for the preparation of a pharmaceutical composition comprising a NK-1 receptor antagonist and an anorectic agent, which process comprises bringing a NK-1 receptor antagonist and an anorectic agent, into association with a pharmaceutically acceptable carrier or excipient.
When administered in combination, either as a single or as separate pharmaceutical composition(s), the NK-1 receptor antagonist and an anorectic agent, are presented in a ratio which is consistent with the manifestation of the desired effect. In particular, the ratio by weight of the NK-1 receptor antagonist and the anorectic agent will suitably be between 0.001 to 1 and 1000 to 1, and especially between 0.01 to 1 and 100 to 1.
A suitable dosage level for the NK-1 receptor antagonist about 0.05 to 1500mg per day, preferably about 0.25 to 1500mg per day, and especially about 0.25 to 500mg/kg per day. The compounds may be administered on a regimen of up to 6 times per day, preferably 1 to 4 times per day, especially 1 or 2 times daily.
A suitable dosage level for the anorectic agent is about 0.5 to 1500mg per day, preferably about 2.5 to lOOOmg per day, and especially about 2.5 to 500mg per day. The compounds may be administered on a regimen of up to 6 times per day, preferably 1 to 4 times per day, especially 1 or 2 times daily.
It will be appreciated that the amount of the NK-1 receptor antagonist and (where present) the anorectic agent required for use in the treatment or prevention of eating disorders will vary not only with the particular compounds or compositions selected but also with the route of administration, the nature of the condition being treated, and the age and condition of the patient, and will ultimately be at the discretion of the patient's physician or pharmacist.
The compounds of formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) may be prepared by the methods described in EP-A-0 577 394 (or WO 95/16679), WO 95/18124, WO 95/23798, WO 96/05181, EP-A-0 436 334, WO 93/21155, EP-A-0 591 040, EP-A-0 532 456, EP-A-0 443 132, WO 92/17449, WO 95/08549 and WO 95/14017, respectively.
Particularly preferred NK-1 receptor antagonists of the formulae (I), (II), (III), (IV), (V), (VI), (VII), (VIII), (IX), (X), (XI) and (XII) for use in the present invention are compounds which are potent NK-1 receptor antagonists, i.e. compounds with an NK-1 receptor affinity (ICso) of less than lOOnM. Even more preferred NK-1 receptor antagonists of use in the present invention are compounds which are potent NK-1 receptor antagonists with an NK-1 receptor affinity (IC50) of less than lOnM, favourably less than 2nM and preferably less than InM. Especially preferred NK-1 receptor antagonists of use in the present invention are orally active, long acting, CNS-penetrant NK-1 receptor antagonists, identified using a combination of the following assays:
ASSAY 1: NK-1 Receptor binding NK-1 receptor binding assays are performed in intact Chinese hamster ovary (CHO) cells expressing the human NK-1 receptor using a modification of the assay conditions described by Cascieri et al, J. Pharmacol. Exp. Ther., 1992, 42, 458. Typically, the receptor is expressed at a level of 3xl05 receptors per cell. Cells are grown in monolayer culture, detached from the plate with enzyme-free dissociation solution (Speciality Media Inc.), and washed prior to use in the assay. i25I-Tyr8-substance P (O.lnM, 2000Ci/mmol; New England Nuclear) is incubated in the presence or absence of test compounds (dissolved in 5μl dimethylsulphoxide, DMSO) with 5xl04 CHO cells. Ligand binding is performed in 0.25ml of 50mM Tris-HCl, pH7.5, containing 5mM MnCl2, 150mM NaCl, 0.02% bovine serum albumin (Sigma), 50μg/ml chymostatin (Peninsula), O.lnM phenylmethylsulphonyl fluoride, 2μg/ml pepstatin, 2μg/ml leupeptin and 2.8μg/ml furoyl saccharine. The incubation proceeds at room temperature until equilibrium is achieved (>40 minutes) and the receptor-ligand complex is harvested by filtration over GF/C filters pre-soaked in 0.1% polyethylenimine using a Tomtek 96-well harvester. Non-specific binding is determined using excess substance P (lμM) and represents <10% of total binding. ASSAY 2: Gerbil Foot-Tapping
CNS-penetrant NK-1 receptor antagonists for use in the present invention can be identified by their ability to inhibit foot tapping in gerbils induced by anxiogenic agents (such as pentagastrin) or central infusion of NK-1 receptor agonists such as GR73632, or caused by aversive stimulation such as foot shock or single housing, based on the method of Rupniak & Williams, Eur. J. Pharmacol, 1994, 265, 179.
Male or female Mongolian gerbils (35-70g) are anaesthetised by inhalation of an isoflurane/oxygen mixture to permit exposure of the jugular vein in order to permit administration of test compounds or vehicle in an injection volume of approximately 5ml/kg i.v. Alternatively, test compounds may be administered orally or by subcutaneous or intraperitoneal routes. A skin incision is then made in the midline of the scalp to expose the skull. An anxiogenic agent (e.g. pentagastrin) or a selective NK-1 receptor agonist (e.g. GR73632 (d Ala[L-Pro9,Me-Leu10]- substance P-(7-ll)) is infused directly into the cerebral ventricles (e.g. 3pmol in 5μl i.c.v., depending on test substance) by vertical insertion of a cuffed 27 gauge needle to a depth of 4.5mm below bregma. The scalp incision is closed and the animal allowed to recover from anaesthesia in a clear perspex observation box (approximately 25cm x 20cm x 20cm). The duration and/or intensity of hind foot tapping is then recorded continuously for approximately 5 minutes. Alternatively, the ability of test compounds to inhibit foot tapping evoked by aversive stimulation, such as foot shock or single housing, may be studied using a similar method of quantification.
ASSAY 3: Ferret Emesis
Individually housed male ferrets (1.0 -2.5 kg) are dosed orally by gavage with test compound. Ten minutes later they are fed with approximately lOOg of tinned cat food. At 60 minutes following oral dosing, cisplatin (lOmg/kg) is given i.v. via a jugular vein catheter inserted under a brief period of halothane anaesthesia. The catheter is then removed, the jugular vein ligated and the skin incision closed. The ferrets recover rapidly from the anaesthetic and are mobile within 10-20 minutes. The animals are observed continuously during recovery from the anaesthetic and for 4 hours following the cisplatin injection, after which time the animals are killed humanely. The numbers of retches and vomits occurring during the 4 hours after cisplatin administration are recorded by trained observers.
ASSAY 4: Separation-Induced Vocalisation
Male and female guinea-pigs pups are housed in family groups with their mothers and littermates throughout the study. Experiments are commenced after weaning when the pups are at least 2 weeks old. Before entering an experiment, the pups may be screened to ensure that a vigorous vocalisation response is reproducibly elicited following maternal separation. The pups are placed individually in an observation cage (approximately 55cm x 39cm x 19cm) in a room physically isolated from the home cage for approximately 15 minutes and the duration and or number of vocalisation during this baseline period is recorded. Those animals which vocalise for longer than 5 minutes are employed for drug challenge studies (approximately 50% of available pups may fail to reach this criterion). On test days each pup receives an oral dose or an s.c. or i.p. injection of test compound or vehicle and is then immediately returned to the home cage with its mother and siblings for at least 30 to 60 minutes (or for up to 4 hours following an oral dose, dependent upon the oral pharmacokinetics of the test compound) before social isolation for 15 minutes as described above. The duration and/or number of vocalisation on drug treatment days may be expressed as a percentage of the pre- treatment baseline value for each animal or compared with values obtained in vehicle-treated animals. The same subjects may be retested once weekly for up to 6 weeks. Between 6 and 8 animals receive each test compound at each dose tested.
A suitable selection cascade for NKi antagonists of use according to the present invention is as follows: (i) Determine affinity for human NKi receptor in radioligand binding studies (Assay 1); select compounds with ICso < lOnM, preferably IC50 < 2nM, especially IC50 < InM.
(ii) Determine ability of compounds to penetrate CNS by their ability to inhibit foot tapping in gerbils induced by central injection of an NKi agonist (Assay 2); select compounds that inhibit foot tapping with ID50 ≤ 3mg/kg i.v., and preferably ID50 < lmg/kg i.v. when administered immediately prior to central NKi agonist challenge, or IDSQ < 30mg/kg p.o., and preferably ID50 ≤ lOmg/kg p.o. 1 hour prior to challenge.
(iii) Determine central duration of action of compounds in gerbil foot tapping assay following intravenous administration 24 hours prior to central NKi agonist challenge; select compounds showing < 25-fold loss of potency compared with IDso determined in step (ii) above with the proviso that ID50 ≤ lOmg/kg i.v., and preferably < 5mg/kg i.v. after 24 hour pre-treatment. (iv) Determine oral bioavailability of compounds by pharmacokinetic analysis, activity in gerbil foot tapping assay following oral administration and/or by ability to inhibit cisplatin-induced emesis in ferrets (Assay 3); select compounds with ID90 < 3mg/kg p.o., and preferably Particularly preferred compounds of use in the present invention are identified using steps (i) to (iv) followed by step (v):
(v) Determine activity of compounds in assays sensitive to conventional serotonergic drugs (inhibition of pharmacologically evoked foot tapping in gerbils and/or inhibition of distress vocalisations in guinea- pig pups (Assay 4)). Select compounds with IDso 20mg/kg, and preferably IDso < lOmg/kg.
Yet further preferred compounds of use in the present invention may be selected from those compounds which satisfy the NK-1 receptor binding criteria of step (i) which, in addition, have < 5-fold shift in affinity when incubated in the presence of human serum albumin (HSA) to show non-specific protein binding.
One example of a NK-1 receptor antagonist of use in the present invention is the compound 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)- ethoxy)-3-(S)-(4-fluorophenyl)-4-(3-(5-oxo-lH,4H-l,2,4-triazolo)methyl)- morpholine, the preparation of which is described in International Patent Specification No. WO 95/16679. In the aforementioned assays, this compound has the following activity:
human NK-1 receptor binding: ICso=0.1nM gerbil foot-tapping (5 mins.): IDso=0.36mg/kg i.v. gerbil foot-tapping (24 hrs.): IDso=0.33mg/kg i.v. ferret emesis: ID9o<3mg/kg p.o. guinea-pig vocalisation IDso=0.73mg/kg p.o.
(4hrs. pretreatment)
Another example of a NK-1 receptor antagonist of use in the present invention is the compound 2-(R)-(l-(R)-(3,5- bis(trifluoromethyl)phenyl)ethoxy)-4-(5-(N,N-dimethylamino)methyl-l,2,3- triazol-4-yl)methyl-3-(S)-phenylmorpholine, the preparation of which is described in International Patent Specification No. WO 95/18124. In the aforementioned assays, this compound has the following activity:
human NK-1 receptor binding: IC5o=0.25nM gerbil foot-tapping (5 mins.): IDso=0.12mg/kg i.v. gerbil foot-tapping (24 hrs.): ID5o=0.17mg/kg i.v. guinea-pig vocalisation: IDso=0.5mg/kg s.c.
The following assay has been used to demonstrate the potentiation of the anorectic effects of dexfenfluramine in diet-induced obese mice when co-administered with a NK-1 receptor antagonist.
Evaluation of the Interaction of NK-1 Antagonists and Anorectic Agents on Food Intake and Body Weight in Diet-Induced Obese Mice.
Mice: Male C57BL/J mice were obtained from Jackson Labs at 3 weeks of age. Half the mice were maintained on a wet diet consisting of sweetened condensed milk and standard ground rodent chow (70%:30%, vol:vol). Fresh wet chow was provided daily. These mice will be referred to as diet- induced obese (DIO). The other half was maintained on just ground rodent chow. These will be referred to as Non-Obese Littermates (NOL). Both food and water were supplied ad libitum. Mice were housed with a 12 hour light/dark cycle (4.00am lights on) through out the course of the described studies.
Mice were weighed bi-weekly until a point that both DIO and NOL mice were weight stable (approximately 20 weeks). At this time, DIO mice weighed significantly more than NOL mice (p20.01). DIO mice also exhibited elevated insulin and glucose levels, as well as polyuria.
Food Intake: (All food intake studies were performed on weight stable DIO mice.
Both food and water were available before treatment.)
The effect that the anorectic agent, dexfenfluramine had on food intake in DIO mice was determined previously (EDso = 3 mg/kg, ip). The combined effect that [2-methoxy-5-(5-trifluoromethyl-tetrazol-l-yl)-benzyl]- ([2S,3S]-2-phenyl-piperidin-3-yl)amine (GR205171) and dexfenfluramine had on food consumption was examined by observing the resulting changes in food intake observed after treatment with dexfenfluramine, GR205171, or combinations of dexfenfluramine with decreasing doses of GR 205171. Mice were randomly assigned to one of the following treatment groups:
• Saline/Saline
• Saline/GR 205171@20 mg/kg
• Dexfenfluramine @ 3 mg/kg/ GR 205171@20 mg/kg
• Dexfenfluramine @ 3 mg/kg/ GR 205171@10 mg/kg • Dexfenfluramine ® 3 mg/kg/ GR 205171® 5 mg/kg
Mice received two injections approximately 30 mins apart. All injections were administered ip., in a volume of 0.2 ml between 3.00pm and 3.30pm. Fresh chow was provided at the time of injection. Food intake was measured 16 hours post-injection for each mouse.
Results shown in Figure 1 are expressed as inhibition of food intake relative to that of saline treated animals.
Body Weight: (All weight studies are performed on DIO mice)
The effect that the combination of anorectic agents and NK-1 antagonists have on weight are examined using a chronic dosing regimen.
Mice are treated with dexfenfluramine, GR 205171, or combinations of dexfenfluramine with decreasing doses of GR 205171, similar to those used in the evaluation of food intake. Mice are dosed once daily, for 7 days with body weights being measured at the start and conclusion of the study.
Changes in body weight are compared with that of saline treated mice. Concurrent daily food intake measurements may be taken at this time. The following examples illustrate pharmaceutical compositions according to the invention.
These formulations may be prepared with separate active ingredients or with a combination of active ingredients in one composition. In such combined preparations, the ratio of the NK-1 receptor antagonist and the anorectic agent will depend upon the choice of active ingredients.
EXAMPLE 1 Tablets containing 50-300mg of NK-1 antagonist and 15mg of dexfenfluramine
Amount mg NK-1 antagonist 50.0 100.0 300.0 dexfenfluramine 15.0 15.0 15.0
Microcrystalline cellulose 80.0 80.0 80.0
Modified food corn starch 80.0 80.0 80.0
Lactose 174.5 124.5 124.5
Magnesium Stearate 0.5 0.5 0.5
EXAMPLE 2 Tablets containing 50-300mg of NK-1 antagonist and 60mg of fenfluramine
Amount mg NK-1 antagonist 50.0 100.0 300.0 fenfluramine 60.0 60.0 60.0
Microcrystalline cellulose 80.0 80.0 80.0
Modified food corn starch 80.0 80.0 80.0
Lactose 129.5 179.5 129.5
Magnesium Stearate 0.5 0.5 0.5
The active ingredients cellulose, lactose and a portion of the corn starch are mixed and granulated with 10% corn starch paste. The resulting granulation is sieved, dried and blended with the remainder of the corn starch and the magnesium stearate. The resulting granulation is then compressed into tablets containing 50mg, lOOmg and 300mg of the NK-1 receptor antagonist per tablet.
EXAMPLE 3 Parenteral iniection
Amount
Active Ingredients 10 to 300mg
Citric Acid Monohydrate 0.75mg
Sodium Phosphate 4.5mg
Sodium Chloride 9mg
Water for injection to 10ml
The sodium phosphate, citric acid monohydrate and sodium chloride are dissolved in a portion of the water. The active ingredients are dissolved or suspended in the solution and made up to volume.

Claims

1. Use of a NK-1 receptor antagonist for the manufacture of a medicament for the treatment or prevention of eating disorders.
2. Use according to claim 1 which further comprises the simultaneous, separate or sequential administration of an anorectic agent.
3. A pharmaceutical composition for the treatment or prevention of eating disorders comprising a NK-1 receptor antagonist, together with at least one pharmaceutically acceptable carrier or excipient.
4. A composition according to claim 3 which further comprises an anorectic agent.
5. A product comprising a NK-1 receptor antagonist and an anorectic agent as a combined preparation for simultaneous, separate or sequential use in the treatment or prevention of eating disorders.
6. A method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment of an effective amount of a NK-1 receptor antagonist.
7. A method for the treatment or prevention of eating disorders, which method comprises administration to a patient in need of such treatment of an amount of a NK-1 receptor antagonist and an amount of an anorectic agent, such that together they give effective relief.
8. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim
6 or 7 wherein the NK-1 receptor antagonist is a compound of formula I:
wherein
Ri is selected from the group consisting of:
(1) Ci-ealkyl, substituted with one or more of the substituents selected from:
(a) heterocycle, wherein the heterocycle is selected from the group consisting of:
(A) benzimidazolyl,
(B) imidazolyl, (C) isoxazolyl,
(D) isothiazolyl,
(E) oxadiazolyl,
(F) pyrazinyl,
(G) pyrazolyl, (H) pyridyl,
(I) pyrrolyl,
(J) tetrazolyl,
(K) thiadiazolyl,
(L) triazolyl, and (M) piperidinyl, and wherein the heterocycle is unsubstituted or substituted with one or more substituent(s) selected from:
(i) Ci-ealkyl, unsubstituted or substituted with halo, -CF3, -OCH3, or phenyl, (ii) Ci-ealkoxy,
(iii) oxo, (iv) thioxo,
(v) cyano,
(vi) -SCH3,
(vii) phenyl, (viii) hydroxy, (ix) trifluoromethyl,
(x) -(CH2)m-NR9R10, wherein m is 0, 1 or 2, and R9 and Rio areindependently selected from:
(I) hydrogen,
(II) Ci-ealkyl,
(III) hydroxyCi-ealkyl, and
(IV) phenyl,
(xi) -NR9CORi┬░, wherein R9 and Rio are as defined above, and
(xii) -CONR9R ┬░, wherein R9 and R o are as defined above, R2 and R3 are independently selected from the group consisting of:
(1) hydrogen;
(2) C╬╣.6alkyl
(3) C2-ealkenyl, and (5) phenyl;
X is -O-; R is
R5 is phenyl, unsubstituted or substituted with halo;
R6, R7 and R8 are independently selected from the group consisting of:
(1) hydrogen,
(2) Ci-ealkyl,
(3) halo, and (4) -CF3; Y is -O-; and
Z is hydrogen or C╬╣- alkyl; or a pharmaceutically acceptable salt thereof.
9. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is a compound of formula Ila:
(Ha) wherein:
A1 is fluorine or CF3;
A2 is fluorine or CF3;
A3 is fluorine or hydrogen;
R6 is a 5-membered or 6-membered heterocyclic ring containing 2 or 3 nitrogen atoms optionally substituted by =O, =S or a C╬╣- alkyl group, and optionally substituted by a group of the formula ZNR7R8 where
Z is Ci-ealkylene or C3-ecycloalkylene;
R7 is hydrogen, C╬╣-4alkyl, C3-7cycloalkyl or C3-7cycloalkylC╬╣-4alkyl, or C2- alkyl substituted by C╬╣.4alkoxy or hydroxyl;
R8 is hydrogen, C╬╣-4alkyl, C3-7cycloalkyl or C3-7cycloalkylC╬╣- alkyl, or C2-4alkyl substituted by one or two substituents selected from C╬╣- alkoxy, hydroxyl or a 4, 5 or 6 membered heteroaliphatic ring containing one or two heteroatoms selected from N, O and S; or R7, R8 and the nitrogen atom to which they are attached form a heteroaliphatic ring of 4 to 7 ring atoms, optionally substituted by a hydroxy group, and optionally containing a double bond, which ring may optionally contain an oxygen or sulphur ring atom, a group S(O) or S(O)2 or a second nitrogen atom which will be part of a NH or NRC moiety where Rc is C╬╣-4alkyl optionally substituted by hydroxy or C╬╣-4alkoxy; or R7, R8 and the nitrogen atom to which they are attached form a non-aromatic azabicyclic ring system of 6 to 12 ring atoms; or Z, R7 and the nitrogen atom to which they are attached form a heteroaliphatic ring of 4 to 7 ring atoms which may optionally contain an oxygen ring atom; or a pharmaceutically acceptable salt thereof.
10. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim
6 or 7 wherein the NK-1 receptor antagonist is a compound of formula III:
wherein:
R2 and R3 are independently selected from the group consisting of:
(1) hydrogen,
(2) Ci-ealkyl,
(3) d-ealkenyl, and (4) phenyl; R6, R7 and R8 are independently selected from the group consisting of:
(1) hydrogen,
(2) Ci-ealkyl,
(3) fluoro, (4) chloro,
(5) bromo,
(6) iodo, and
(7) -CF3;
Ru, Ri2 and Ri3 are independently selected from the group consisting of: (1) fluoro,
(2) chloro,
(3) bromo, and
(4) iodo;
A is unsubstituted l-ealkyl; B is selected from the group consisting of:
p is 0 or 1;
X is selected from:
(a) -PO(OH)O" ΓÇó M+, wherein M+ is a pharmaceutically acceptable monovalent counterion,
(b) -PO(O-)2 ΓÇó 2M+,
(c) -PO(O")2 ΓÇó D2+, wherein D2+ is a pharmaceutically acceptable divalent counterion,
(d) -CH(R )-PO(OH)O- ΓÇó M+, wherein R4 is hydrogen or Ci-salkyl, (e) -CH(R )-PO(O")2 ΓÇó 2M+,
(f) -CH(R4)-PO(O-)2 ΓÇó D +,
(i) -CO-CH2CH2-CO2- ΓÇó M+,
(j) -CH(CH3)-O-CO-R5, wherein R5 is selected from the group consisting of:
(i) , NH3 M" " 0
HΓÇ₧ M"
(ϋ) .N?
O ' OH
(ϋi) 0 C02 ' M
C02 " M"
(iv)
0 C02 " M"
C00
(v) 0
NHQ
Y is -O-;
Z is hydrogen or Ci-ealkyl; or a pharmaceutically acceptable salt thereof.
11. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is a compound of formula IVa:
wherein
Ai is fluorine or CF3; A2 is fluorine or CF3;
A3 is fluorine or hydrogen;
X is a group of the formula NR6R7 or a C- or N-linked imidazolyl ring;
Y is hydrogen or C╬╣-4alkyl optionally substituted by a hydroxy group;
RG is hydrogen, Ci-Galkyl, C3-7cycloalkyl, C3-7cycloalkylC╬╣-4alkyl, phenyl, or C2-4alkyl substituted by C╬╣-4alkoxy or hydroxy;
R7 is hydrogen, Ci-ealkyl, C3-7cycloalkyl, C3-7cycloalkylC╬╣-4alkyl, phenyl, or C2-4alkyl substituted by one or two substituents selected from C╬╣-4alkoxy, hydroxy or a 4, 5 or 6 membered heteroaliphatic ring containing one or two heteroatoms selected from N, O and S; or R6 and R7, together with the nitrogen atom to which they are attached, form a saturated or partially saturated heterocyclic ring of 4 to 7 ring atoms, which ring may optionally contain in the ring one oxygen or sulphur atom or a group selected from NR8, S(O) or S(0)2 and which ring may be optionally substituted by one or two groups selected from hydroxyC╬╣- alkyl, C╬╣- alkoxyC╬╣- alkyl, oxo, CORa or CO2Ra where Ra is hydrogen or C╬╣-4alkyl; or RG and R7 together with the nitrogen atom to which they are attached, form a non-aromatic azabicyclic ring system of 6 to 12 ring atoms; and
R8 is hydrogen, C╬╣-4alkyl, hydroxy C╬╣-4alkyl or C╬╣. alkoxyC╬╣-4alkyl; or a pharmaceutically acceptable salt thereof.
12. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is a compound of formula V:
R7
wherein:
Y is (CH2)n wherein n is an integer from 1 to 4, and wherein any one of the carbon-carbon single bonds in said (CH2)n may optionally be replaced by a carbon-carbon double bond, and wherein any one of the carbon atoms of said (CH2)n may optionally be substituted with R4, and wherein any one of the carbon atoms of said (CH2)n may optionally be substituted with R7;
Z is (CH2)m wherein m is an integer from 0 to 6, and wherein any one of the carbon-carbon single bonds of (CH2)m may optionally be replaced by a carbon-carbon double bond or a carbon-carbon triple bond, and any one of the carbon atoms of said (CH2)m may optionally be substituted with R8;
Ri is hydrogen or Ci-salkyl optionally substituted with hydroxy, C╬╣-4alkoxy or fluoro; R2 is a radical selected from hydrogen, Ci-e straight or branched alkyl, C3-7cycloalkyl wherein one of the CH2 groups in said cycloalkyl may optionally be replaced by NH, oxygen or sulphur; aryl selected from phenyl and naphthyl; heteroaryl selected from indanyl, thienyl, furyl, pyridyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl and quinolyl; phenyl-C2-6alkyl, benzhydryl and benzyl, wherein each of said aryl and heteroaryl groups and the phenyl moieties of said benzyl, phenyl - C2-ealkyl and benzhydryl may optionally be substituted with one or more substituents independently selected from halo, nitro, Ci-e alkyl, Ci-ealkoxy, trifluoromethyl, amino, Ci-ealkylamino, Ci-ealkyl-O-CO, Ci-ealkyl-O-CO-
Ci-ealkyl, Ci-ealkyl-CO-O, d-ealkyl-CO-Ci-ealkyl-O-, Ci-ealkyl-CO, Ci-ealkyl-CO-Ci-ealkyl-, di-Ci-ealkylamino, -CONH-Ci-ealkyl, Ci-ealkyl-CO-NH-Ci.ealkyl, -NHCOH and -NHCO-Ci-ealkyl; and wherein one of the phenyl moieties of said benzhydryl may optionally be replaced by naphthyl, thienyl, furyl or pyridyl;
R5 is hydrogen, phenyl or Ci-ealkyl; or R2 and R5 together with the carbon to which they are attached, form a saturated ring having from 3 to 7 carbon atoms wherein one of the CH2 groups in said ring may optionally be replaced by oxygen, NH or sulfur;
R3 is aryl selected from phenyl and naphthyl; heteroaryl selected from indanyl, thienyl, furyl, pyridyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, tetrazolyl and quinolyl; and cycloalkyl having 3 to 7 carbon atoms wherein one of the (CH2) groups in said cycloalkyl may optionally be replaced by NH, oxygen or sulphur; wherein each of said aryl and heteroaryl groups may optionally be substituted with one or more substituents, and said C3-7cycloalkyl may optionally be substituted with one or two substituents, each of said substituents being independently selected from halo, nitro, Ci-ealkyl, Ci-ealkoxy, trifluoromethyl, amino, Ci-ealkylamino, -CO-NH- Ci-ealkyl,
C╬╣-calkyl-CO-NH-C╬╣-ealkyl, -NHCOH and -NHCO-Ci-ealkyl; R4 and R7 are each independently selected from hydroxy, halogen, halo, amino, oxo, cyano, methylene, hydroxymethyl, halomethyl, C╬╣-6alkylamino, di-C╬╣-6alkylamino, Ci-ealkoxy, Ci-ealkyl-O-CO, Ci-ealkyl-O-CO-Ci-ealkyl, Ci-ealkyl-CO-O, Ci-ealkyl-CO-Ci-ealkyl-O-, C╬╣-6alkyl-CO-, C╬╣-6alkyl-CO-C╬╣-6alkyl, and the radicals set forth in the definition of R2;
Ro is -NHCOR9, -NHCH2R9, SO2R8 or one of the radicals set forth in any of the definitions of R2, R4 and R7;
R8 is oximino (=NOH) or one of the radicals set forth in any of the definitions of R2, R4 and R7;
R9 is Ci-╬▓alkyl, hydrogen, phenyl or phenylCi-ealkyl; with the proviso that (a) when m is 0, R8 is absent, (b) when R4, RG, R7 or R8 is as defined in R2, it cannot form together with the carbon to which it is attached ,a ring with R5, and (c) when R4 and R7 are attached to the same carbon atom, then either each of R4 and R7 is independently selected from hydrogen, fluoro and Ci-╬▓alkyl, or R4 and R7, together with the carbon to which they are attached, for a C3-6 saturated carbocyclic ring that forms a spiro compound with the nitrogen-containing ring to which they are attached; or a pharmaceutically acceptable salt thereof.
13. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is a compound of formula VI:
wherein: radicals R are phenyl radicals optionally 2- or 3-substituted by a halogen atom or a methyl radical;
R1 is optionally substituted phenyl, cyclohexadienyl, naphthyl, indenyl or optionally substituted heterocycle; R2 is H, halogen, OH, alkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, alkyloxy, alkylthio, acyloxy, carboxy, optionally substituted alkyloxycarbonyl, benzyloxycarbonyl, amino or acylamino;
R3 is optionally 2-substituted phenyl;
R4 is OH or fluorine when R5 is H; or R4 and R5 are OH ; or R4 and R5 together form a bond; or a pharmaceutically acceptable salt thereof.
14. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim
6 or 7 wherein the NK-1 receptor antagonist is a compound of formula VII:
Ar-T- CO-N- CH2 -C-CH2 - CH2 -Am + , A ~ (VII)
Ar ' wherein:
Ar represents an optionally substituted mono-, di- or tricyclic aromatic or heteroaromatic group;
T represents a bond, a hydroxymethylene group, a Cι. alkoxymethylene group or a Ci-salkylene group; Ar' represents a phenyl group which is unsubstituted or substituted by one or more substituents selected from halogen, preferably chlorine or fluorine, trifluoromethyl, Cι- alkoxy, Cι- alkyl where the said substituents may be the same or different; a thienyl group; a benzothienyl group; a naphthyl group; or an indolyl group; R represents hydrogen, Cι-4alkyl, ω-Cι-4alkoxyCι-4alkyl, or ω-C2-4alkanoyloxyC2-4alkyl; represents hydrogen; or Q and R together form a 1,2-ethylene, 1,3-propylene or 1,4- butylene group;
Am+ represents the radical
in which Xi, X2 and X3, together with the nitrogen atom to which they are attached, form an azabicyclic or azatricyclic ring system optionally substituted by a phenyl or benzyl group; and
A- represents a pharmaceutically acceptable anion.
15. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is a compound of formula VIII
wherein:
Ri represents an optionally substituted aralkyl, aryloxyalykl, heteroaralkyl, aroyl, heteroaroyl, cycloalkylcarbonyl, aralkanoyl, heteroarylalkanoyl, aralkoxycarbonyl or arylcarbamoyl group or the acyl group of an ╬▒-amino acid optionally N-substituted by a lower alkanoyl or carbamoyl-lower alkanoyl group;
R2 represents cycloalkyl or an optionally substituted aryl or heteroaryl group; R3 represents hydrogen, alkyl, carbamoyl or an alkanoyl or alkenoyl group optionally substituted by carboxy or esterified or amidated carboxy; R4 represents an optionally substituted aryl group or an optionally partially saturated heteroaryl group;
Xi represents methylene, ethylene, a bond, an optionally ketalised carbonyl group or an optionally etherified hydroxymethylene group;
X2 represents alkylene, carbonyl or a bond; and
X3 represents carbonyl, oxo-lower alkyl, oxo(aza)-lower alkyl, or an alkyl group optionally substituted by phenyl, hydroxymethyl, optionally esterified or amidated carboxy, or (in other than the ╬▒-position) hydroxy; or a pharmaceutically acceptable salt thereof.
16. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is a compound of formula IX:
wherein:
Ri is aryl, or a group of the formula:
X is CH or N; and
Z is O or N-R5, in which R5 is hydrogen or lower alkyl;
R2 is hydroxy or lower alkoxy;
R3 is hydrogen or optionally substituted lower alkyl;
R4 is optionally substituted ar(lower)alkyl;
A is carbonyl or sulfonyl; and Y is a bond or lower alkenylene; or a pharmaceutically acceptable salt thereof.
17. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is a compound of formula X:
wherein: Ri is aryl selected from indanyl, phenyl and naphthyl; heteroaryl selected from thienyl, furyl, pyridyl and quinolyl; and cycloalkyl having 3 to 7 carbon atoms, wherein one of said carbon atoms may optionally be replaced by nitrogen, oxygen or sulfur; wherein each of said aryl and heteroaryl groups may optionally be substituted with one or more substituents, and said C3-7cycloalkyl may optionally be substituted with one or two substituents, said substituents being independently selected from chloro, fluoro, bromo, iodo, nitro, Ci-ioalkyl optionally substituted with from one to three fluoro groups, Ci-ioalkoxy optionally substituted with from one to three fluoro groups, amino, C╬╣-╬╣oalkyl-S-, C╬╣-╬╣oalkyl-S(O)-, C╬╣.╬╣oalkyl-S╬╕2-; phenyl, phenoxy, C╬╣-╬╣0alkyl-SO2NH-,
C╬╣-╬╣oalkyl-SO2NH-C╬╣-╬╣oakyl-, d-╬╣oalkylamino-diC╬╣-╬╣oalkyl-, cyano, hydroxy, cycloalkoxy having 3 to 7 carbon atoms, Ci-ealkylamino, Ci-edialkylamino, HC(O)NH- and C╬╣-╬╣oalkyl-C(0)NH-; and
R2 is thienyl, benzhydryl, naphthyl or phenyl optionally substituted with from one to three substituents independently selected from chloro, bromo, fluoro, iodo, cycloalkoxy having 3 to 7 carbon atoms, Ci-ioalkyl optionally substituted with from one to three fluoro groups and Ci-ioalkoxy optionally substituted with from one to three fluoro groups; or a pharmaceutically acceptable salt thereof.
18. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is a compound of formula XI:
wherein:
Ri is a C╬╣-4alkoxy group; R2 is
R3 is a hydrogen or halogen atom;
R4 and R5 may each independently represent a hydrogen or halogen atom, or a C╬╣- alkyl, C╬╣- alkoxy or trifluoromethyl group;
R6 is a hydrogen atom, a C╬╣-4alkyl, (CH2)mcyclopropyl, -S(0)nC╬╣- 4alkyl, phenyl, NR7R8, CH2C(O)CF3 or trifluoromethyl group;
R7 and R8 may each independently represent a hydrogen atom, or a C╬╣- alkyl or acyl group; x represents zero or 1; n represents zero, 1 or 2; and m represents zero or 1; or a pharmaceutically acceptable salt thereof.
19. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is a compound of formula XII:
R8 R4
RΓÇö (CH2)nΓÇö C-CHΓÇö NΓÇö (CH2)oΓÇö R3 NH R
I
(CO)p
wherein: m is zero, 1, 2 or 3; n is zero or 1; o is zero, 1 or 2; p is zero or 1;
R is phenyl, 2- or 3-indolyl, 2- or 3-indolinyl, benzothienyl, benzofuranyl, or naphthyl; which R groups may be substituted with one or two halo, C╬╣-3alkoxy, trifluoromethyl, C╬╣-4alkyl, phenyl-C╬╣-3alkoxy, or C╬╣- alkanoyl groups;
R1 is trityl, phenyl, diphenylmethyl, phenoxy, phenylthio, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, indolinyl, indolyl, benzothienyl, hexamethyleneimmyl, benzofuranyl, tetrahydropyridinyl, quinolinyl, isoquinolinyl, reduced quinolinyl, reduced isoquinolinyl, phenyl-(C╬╣-4alkyl)-, phenyl-(C╬╣. alkoxy)-, quinolinyl-(C╬╣-4alkyl)-, isoquinolinyl-(C╬╣-4alkyl)-, reduced quniolinyl-(C╬╣. alkyl)-, reduced isoquinolinyl-(C╬╣- alkyl)-, benzoyl-(C╬╣-3alkyl)-, C╬╣. alkyl, or -NH-CH2-R5; any one of which Ri groups may be substituted with halo, C╬╣- alkyl, C╬╣-4alkoxy, trifluoromethyl, amino, C╬╣-4alkylamino, di(C╬╣-4alkyl)amino, or C2-4alkanoylamino ; or any one of which R groups may be substituted with phenyl, piperazinyl, C3-8cycloalkyl, benzyl, C╬╣-4alkyl, piperidinyl, pyridinyl, pyrimidinyl, C2-6alkanoylamino, pyrrolidinyl, d ealkanoyl, or C╬╣-4alkoxycarbonyl; any one of which groups may be substituted with halo, C╬╣-4alkyl, C╬╣-4alkoxy, trifluoromethyl, amino, C╬╣-4alkylamino, di(C╬╣-4alkyl)amino, or C2-4alkanoylamino; or R1 is amino, a leaving group, hydrogen, C╬╣. alkylamino, or di(C╬╣-4alkyl)amino;
R5 is pyridyl, anilino-(C╬╣-3alkyl)-, or anilinocarbonyl;
R2 is hydrogen, C╬╣-4alkyl, C╬╣. alkylsulfonyl, carboxy-(C╬╣-3alkyl)-, C╬╣-3alkoxycarbonyl-(C╬╣-3alkyl)-, or -CO-R6;
RG is hydrogen, C╬╣-4alkyl, C╬╣.3haloalkyl, phenyl, Ci-3alkoxy, C╬╣-3hydroxyalkyl, amino, C╬╣-4alkylamino, di(C╬╣-4alkyl) amino, or -(CH2VR7; q is zero to 3;
R7 is carboxy, C╬╣-4alkoxycarbonyl, C╬╣- alkylcarbonyloxy, amino, C╬╣- alkylamino, di(C╬╣- alkyl)amino, C╬╣-6alkoxycarbonylamino, or phenoxy, phenylthio, piperazinyl, piperidinyl, pyrrolidinyl, morpholinyl, indolinyl, indolyl, benzothienyl, benzofuranyl, quinolinyl, phenyl-(C╬╣.4alkyl)-, quinolinyl-(C╬╣- alkyl)-, isoquinolinyl-(C╬╣- alkyl)-, reduced quinolinyl- (C╬╣-4alkyl)-, reduced isoquinolinyl-(C╬╣-4alkyl)-, benzoyl-C╬╣-3alkyl; any one of which aryl or heterocyclic R7 groups may be substituted with halo, trifluoromethyl, C╬╣-4alkoxy, C╬╣-4alkyl, amino, C╬╣-4alkylamino, di(C╬╣-4alkyl)amino, or C2-4alkanoylamino; or any one of which R7 groups may be substituted with phenyl, piperazinyl, d-scycloalkyl, benzyl, piperidinyl, pyridinyl, pyrimidinyl, pyrrolidinyl, C2-6alkanoyl, or C╬╣-4alkoxycarbonyl; any of which groups may be substituted with halo, trifluoromethyl, amino, C╬╣-4alkoxy, C╬╣-4alkyl, C╬╣-4alkylamino, di(C╬╣- alkyl)amino, or C2-4alkanoylamino;
R8 is hydrogen or Ci-╬▓alkyl; R3 is phenyl, phenyl-(C╬╣-6alkyl)-, d-╬▓cycloalkyl, Cs-scycloalkenyl,
Ci-salkyl, naphthyl, d-salkenyl, or hydrogen; any one or which groups except hydrogen may be substituted with one or two halo, C╬╣-3alkoxy, d-3alkylthio, nitro, trifluoromethyl, or C╬╣-3alkyl groups; and R4 is hydrogen or C╬╣-3alkyl; with the proviso that if Ri is hydrogen or halo, R3 is phenyl, phenyl-(C╬╣-6alkyl)-, C3-8cycloalkyl, Cs-scycloalkenyl, or naphthyl; or a pharmaceutically acceptable salt thereof.
20. A use, composition, product or method according to any one of the preceding claims wherein the NK-1 receptor antagonist is orally active, long acting and CNS-penetrant.
21. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim
6 or 7 wherein the NK-1 receptor antagonist is selected from the classes of compounds described in EP-A-0577394, WO-A-9508549, WO-A-9518124, WO-A-9523798 and WO-A-9605181.
22. A use according to claim 1 or 2, a composition according to claim 3 or 4, a product according to claim 5 or a method according to claim 6 or 7 wherein the NK-1 receptor antagonist is selected from 2-(S)-(3,5-bis(trifluoromethyl)benzyloxy)-3(S)-(4-fluorophenyl)-4-(3-(5-oxo- lH,4H-l,2,4-triazolo)methyl)morpholine; 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(3-(5-oxo-lH,4H- l,2,4-triazolo)methyl)-3-(S)-phenyl-morpholine; 2-(S)-(3,5-bis(trifluoromethyl)benzyloxy)-4-(3-(5-oxo-lH,4H-l,2,4- triazolo)methyl)-3-(S)-phenyl-morpholine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-
4-(3-(5-oxo-lH,4H-l,2,4-triazolo)methyl)morpholine; 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(5-(N,N- dimethylamino)methyl-l, 2, 3-triazol-4-yl)methyl-3-(S)-phenylmorp holine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(5-(N,N- dimethylamino)methyl-l,2,3-triazol-4-yl)methyl-3-(S)-(4- fluorophenyl)morp holine; 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-
4-(3-(4-monophosphoryl-5-oxo-lH-l,2,4-triazolo)methyl)morpholine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-
4-(3-(l-monophosphoryl-5-oxo-lH-l, 2, 4-triazolo)methyl)morp holine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)- 4-(3-(2-monophosphoryl-5-oxo-lH-l, 2, 4-triazolo)methyl)morp holine;
2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-
4-(3-(5-oxyphosphoryl-lH-l,2,4-triazolo)methyl)morpholine;
2-(S)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-3-(S)-(4-fluorophenyl)-
4-(3-(l-monophosphoryl-5-oxo-4H-l, 2, 4-triazolo)methyl)morp holine; 2-(R)-(l-(R)-(3,5-bis(trifluoromethyl)phenyl)ethoxy)-4-(4-N,N- dimethylaminobut- 2-y n-yl) - 3- (S) - (4-fluorop he nyl)morp holine ; or a pharmaceutically acceptable salt thereof.
23. A use, composition, product or method according to any one of the preceding claims wherein the anorectic agent is selected from aminorex, amphechloral, amphetamine, benzphetamine, chlorphentermine, clobenzorex, cloforex, clominorex, clortermine, cyclexedrine, dexfenfluramine, dextroamphetamine, diethylpropion, diphemethoxidine, N-ethylamphetamine, fenbutrazate, fenfluramine, fenisorex, fenproporex, fludorex, fluminorex, furfurylmethylamphetamine, levamfetamine, levophacetoperane, mazindol, mefenorex, metamfepramone, methamphetamine, norpseudoephedrine, pentorex, phendimetrazine, phenmetrazine, phentermine, phenylpropanolamine, picilorex and sibutramine; or a pharmaceutically acceptable salt thereof.
24. A use, composition, product or method according to any one of the preceding claims wherein the anorectic agent is a halogenated amphetamine derivative.
25. A use, composition, product or method according to claim 24 wherein the amphetamine derivative is selected from chlorphentermine, cloforex, clortermine, dexfenfluramine, fenfluramine, picilorex and sibutramine; or a pharmaceutically acceptable salt thereof.
26. A use, composition, product or method according to any one of claims 1 to 25 wherein the eating disorder is obesity.
27. A use, composition, product or method according to any one of claims 1 to 25 wherein the eating disorder is bulimia nervosa.
28. A use, composition, product or method according to any one of claims 1 to 25 wherein the eating disorder is a compulsive eating disorder.
EP98917425A 1997-04-24 1998-04-22 Use of nk-1 receptor antagonists for treating eating disorders Withdrawn EP0977572A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GBGB9708289.5A GB9708289D0 (en) 1997-04-24 1997-04-24 Therapeutic use
GB9708289 1997-04-24
GB9721265 1997-10-07
GBGB9721265.8A GB9721265D0 (en) 1997-10-07 1997-10-07 Therapeutic use
PCT/GB1998/001161 WO1998047513A1 (en) 1997-04-24 1998-04-22 Use of nk-1 receptor antagonists for treating eating disorders

Publications (1)

Publication Number Publication Date
EP0977572A1 true EP0977572A1 (en) 2000-02-09

Family

ID=26311439

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98917425A Withdrawn EP0977572A1 (en) 1997-04-24 1998-04-22 Use of nk-1 receptor antagonists for treating eating disorders

Country Status (5)

Country Link
EP (1) EP0977572A1 (en)
JP (1) JP2001524960A (en)
AU (1) AU744261B2 (en)
CA (1) CA2287487A1 (en)
WO (1) WO1998047513A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL145240A (en) * 1999-03-19 2007-02-11 Abbott Gmbh & Co Kg A compound and the use thereof for treating eating disorders
JP2003511410A (en) * 1999-10-13 2003-03-25 グラクソ グループ リミテッド Morpholinol derivatives for the treatment of obesity
AU2002247367B2 (en) * 2001-03-21 2005-10-27 Pharmacopeia Drug Discovery, Inc. Aryl and biaryl compounds having MCH modulatory activity
DE10311984A1 (en) * 2003-03-12 2004-09-23 Freie Universität Berlin Using neutral endopeptidase-associated molecules for treatment, diagnosis, prophylaxis and monitoring of eating and metabolic disorders and dementia, also for drug development
CN106290894A (en) * 2016-07-21 2017-01-04 浙江理工大学 A kind of leukemia detection kit based on NK1R albumen

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8929070D0 (en) * 1989-12-22 1990-02-28 Fujisawa Pharmaceutical Co Peptide compounds,processes for preparation thereof and pharmaceutical composition comprising the same
WO1991009844A1 (en) * 1990-01-04 1991-07-11 Pfizer Inc. Substance p antagonists
CA2106200C (en) * 1991-03-26 1996-11-19 Terry J. Rosen Stereoselective preparation of substituted piperidines
MY110227A (en) * 1991-08-12 1998-03-31 Ciba Geigy Ag 1-acylpiperindine compounds.
FR2689888B1 (en) * 1992-04-10 1994-06-10 Rhone Poulenc Rorer Sa NOVEL PERHYDROISOINDOLE DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
IL106142A (en) * 1992-06-29 1997-03-18 Merck & Co Inc Morpholine and thiomorpholine tachykinin receptor antagonists, their preparation and pharmaceutical compositions containing them
FR2696178B1 (en) * 1992-09-30 1994-12-30 Sanofi Elf Quaternary basic amides, process for their preparation and pharmaceutical compositions containing them.
IS4208A (en) * 1993-09-22 1995-03-23 Glaxo Group Limited 3- (tetrazolyl-benzyl) amino-piperadidine derivatives
US6403577B1 (en) * 1993-11-17 2002-06-11 Eli Lilly And Company Hexamethyleneiminyl tachykinin receptor antagonists
CZ288176B6 (en) * 1993-12-29 2001-05-16 Merck Sharp & Dohme Substituted morpholine derivatives, process of their preparation, their use for preparing pharmaceutical preparations and pharmaceutical preparations in which they are comprised
IL112778A0 (en) * 1994-03-04 1995-05-26 Merck & Co Inc Substituted heterocycles, their preparation and pharmaceutical compositions containing them
PT777659E (en) * 1994-08-15 2001-09-28 Merck Sharp & Dohme MORPHINE DERIVATIVES AND THEIR USE AS THERAPEUTIC AGENTS
WO1996024353A1 (en) * 1995-02-10 1996-08-15 Eli Lilly And Company Methods of treating or preventing psychiatric disorders
EP0839043A2 (en) * 1995-05-25 1998-05-06 BioFrontiers, Inc. Pharmaceutical compositions containing calcium sulfate
ZA966891B (en) * 1995-08-21 1998-02-16 Lilly Co Eli 2-acylaminopropanamines as growth hormone secretagogues.
AU2611297A (en) * 1996-04-12 1997-11-07 Eli Lilly And Company Bisindoles for treating pain or nociception
AU722541B2 (en) * 1996-06-21 2000-08-03 Merck Sharp & Dohme Limited Spiro-piperidine derivatives and their use as therapeutic agents
GB9613969D0 (en) * 1996-07-03 1996-09-04 Merck Sharp & Dohme Therapeutic agents

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9847513A1 *

Also Published As

Publication number Publication date
WO1998047513A1 (en) 1998-10-29
AU744261B2 (en) 2002-02-21
JP2001524960A (en) 2001-12-04
AU7065798A (en) 1998-11-13
CA2287487A1 (en) 1998-10-29

Similar Documents

Publication Publication Date Title
US6162805A (en) Use of an NK-1 receptor antagonist and an SSRI for treating obesity
US6232311B1 (en) Method for treating or preventing psychosomatic and psychoimmunologic disorders
EP0977572A1 (en) Use of nk-1 receptor antagonists for treating eating disorders
US6090819A (en) Use of NK-1 receptor antagonists for treating mania
AU749976B2 (en) Method for treating premenstrual or late luteal phase syndrome
AU737019B2 (en) Use of NK-1 receptor antagonists for treating aggressive behaviour disorders
AU745524B2 (en) Method for treating attention deficit disorder
AU738047B2 (en) Use of NK-1 receptor antagonists for treating mania
WO1999027938A1 (en) Method for treating or preventing psychosomatic and psychoimmunologic disorders
US20020142942A1 (en) Method for treating premenstrual or late luteal phase syndrome
WO1999007374A1 (en) Use of nk-1 receptor antagonists for treating aggressive behaviour
EP1001780A1 (en) Use of nk-1 receptor antagonists for treating mania
EP1001779A1 (en) Use of nk-1 receptor antagonists for treating aggressive behaviour disorders
EP1085874A1 (en) Use of a nk-1 receptor antagonist for treating sexual dysfunctions
JP2001513500A (en) Use of an NK-1 receptor antagonist for the treatment of mania
JP2001513499A (en) Use of a NK-1 receptor antagonist for the treatment of aggressive behavioral disorders

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19991124

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU NL PT SE

17Q First examination report despatched

Effective date: 20010201

GRAH Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOS IGRA

RIC1 Information provided on ipc code assigned before grant

Free format text: 7A 61K 31/535 A, 7A 61K 31/675 B, 7A 61K 31/435 B, 7A 61K 31/445 B, 7A 61K 31/40 B, 7A 61K 31/47 B, 7A 61P 3/04 B

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20030225