EP1107748A2 - Procedes et compositions employant le vigabatrin s(+) optiquement pur - Google Patents

Procedes et compositions employant le vigabatrin s(+) optiquement pur

Info

Publication number
EP1107748A2
EP1107748A2 EP99945177A EP99945177A EP1107748A2 EP 1107748 A2 EP1107748 A2 EP 1107748A2 EP 99945177 A EP99945177 A EP 99945177A EP 99945177 A EP99945177 A EP 99945177A EP 1107748 A2 EP1107748 A2 EP 1107748A2
Authority
EP
European Patent Office
Prior art keywords
vigabatrin
administered
pharmaceutically acceptable
acceptable salt
amount
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Withdrawn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
EP99945177A
Other languages
German (de)
English (en)
Inventor
Paul D. Rubin
Timothy J. Barberich
William E. Yelle
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.)
Sunovion Pharmaceuticals Inc
Original Assignee
Sepracor Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sepracor Inc filed Critical Sepracor Inc
Publication of EP1107748A2 publication Critical patent/EP1107748A2/fr
Withdrawn legal-status Critical Current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
    • A61K31/435Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid or pantothenic acid
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse

Definitions

  • the invention relates to methods of prevention, treatment, and/or management of peripheral neuropathy or the symptoms thereof, drug addiction or the symptoms thereof, and symptoms associated with drug withdrawal.
  • Peripheral neuropathy in general is a syndrome that can occur upon the disease or damage of nervous tissue. It is characterized, alone or in combination, by sensory loss, muscle weakness and atrophy, decreased deep tendon reflexes, and vasomotor symptoms. Peripheral neuropathies are classified as those that occur upon disease or damage of a single nerve (mononeuropathy), upon disease or damage of two or more nerves in separate areas (multiple mononeuropathy), and upon disease or damage of many nerves simultaneously (polyneuropathy). Peripheral neuropathies may also be classified by the general type of nerve(s) affected. For example, some neuropathies involve motor fibers, whereas others mainly affect sensory fibers (dorsal root ganglia) or cranial nerves. Some relatively common peripheral neuropathies have also been given specific names: carpal tunnel syndrome is one example; Guillain-Barre syndrome is another.
  • Carpal tunnel syndrome is an affliction endured by millions. Occurring most often in women, it is particularly associated with occupations that require repeated forceful wrist flexion, as well as with acromegaly, myxedema, and fluid changes of pregnancy. CTS results from compression of the median nerve in the volar aspect of the wrist between the longitudinal tendons of forearm muscles that flex the hand and the transverse superficial carpal ligament. This compression causes pain, sensory deficit in the palmar aspect of the first three digits, and/or weakness and atrophy in muscles controlling thumb movement. The Merck Manual, 16th ed., 1519 ( 1992). Guillain-Barre syndrome (GBS) is another specific kind of peripheral neuropathy.
  • GBS is an acute, usually rapidly progressive form of inflammatory polyneuropathy characterized by muscular weakness and mild distal sensory loss. It typically occurs between about 5 days to 3 weeks after a banal infectious disorder, surgery, or an immunization, and is thus believed to be caused by an autoimmune reaction of some kind. The most frequently identified cause of GBS is Campylobacter jejuni infection. Hughes, R. A. C, et al, J. Infect. Diseases, 176(Suppl. 2):S92-8 (1997).
  • the diseases and injuries that can lead to peripheral neuropathies generally are numerous and varied. They include, but are not limited to: trauma caused by injury or surgical operation; tumors; bony hyperostosis; casts; crutches; prolonged cramped postures; hemorrhage into a nerve; exposure to cold or radiation; collagen-vascular disorders; metabolic diseases such as diabetes; infectious diseases such as Lyme disease and HTV; toxins such as emetine, hexobarbital, barbital, chlorobutanol, sulfonamides, phenytoin, nitrofurantoin, the vinca alkaloids, heavy metals, carbon monoxide, triorthocresylphosphate, orthodinitrophenol, and other solvents and industrial poisons; autoimmune reactions; nutritional deficiency and vitamin B deficiency in particular; and metabolic disorders such as hypothyroidism, porphyria, sarcoidosis, amyloidosis, uremia and diabetes.
  • peripheral neuropathy The nature and effectiveness of conventional treatments of peripheral neuropathy vary with the underlying disorder. For example, successful renal transplantation usually diminishes the uremic polyneuropathy that occurs in about half of patients undergoing dialysis. Pirzada, N. A., et al, Postgrad. Med., 102:249-261 (1997). Similarly, immunologically mediated neuropathy may be treated with immunosuppression or immunomodulation therapy. Openshaw, H., Biol. Blood and Marrow Trans., 3:202-209 (1997). Because the etiology of peripheral neuropathy is often difficult to determine, however, its treatment is often slow and ineffective. See, e.g., Hughes, R. A. C, et al., J. Infect.
  • Vigabatrin chemically named 4-amino-5-hexanoic acid
  • GABA-T gamma-aminobutyric acid-transaminase
  • GABA-T inactivates the inhibitory neurotransmitter 4-aminobutyric acid (GABA), which acts at the brain's GABA/benzodiazepine receptor to increase membrane conductance of chloride ions, stabilizing the membrane potential, and consequently dampening excitatory input.
  • vigabatrin By inhibiting the metabolism of GABA, vigabatrin can increase GABA concentrations in the central nervous system to levels which suppress or diminish the intensity of a variety of seizures. Because vigabatrin is not degraded by the GABA-T enzyme, only new enzyme synthesis can restore GABA-T activity. Thus, even a single dose produces a lasting, dose-dependent inhibition of brain
  • GABA-T in experimental animals, and elevated GABA levels are seen for at least 24 hours. Shields, W. D., and Sankar, R., Sem. Ped. Neur., 4(l):43-50 (1997).
  • vigabatrin is currently used in other countries as an anti-epileptic agent. Shields, W. D., and Sankar, R., Sem. Ped. Neur., 4(l):43-50 (1997). It is commonly available as a racemic mixture of the R(-) and S(+) stereoisomers. Further, the S(+) stereoisomer is reported to be active as a GABA-T inhibitor, and in in vivo animal studies of anti-convulsant activity. Gram, L., et al, Brit. J. Clinical Pharmacol, 27:13S-18S, (1989).
  • vigabatrin for the treatment of a variety of conditions and disorders, particularly in a dosage form that is convenient for administration to a patient, without any of the known disadvantages of vigabatrin.
  • This invention further encompasses a method of treating or preventing peripheral neuropathy which comprises administering to a mammal, preferably a human, in need of such treatment a therapeutically effective amount of S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer.
  • the invention encompasses a method of treating or preventing carpal tunnel syndrome which comprises administering to a mammal, preferably a human, in need of such treatment a therapeutically effective amount of S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer.
  • This invention further encompasses a method of treating or preventing Guillain-Barre syndrome which comprises administering to a mammal, preferably a human, in need of such treatment a therapeutically effective amount of S(+) vigabatrin, or a pharmaceutically acceptable salt thereof substantially free of its R(-) stereoisomer.
  • This invention also encompasses a method of preventing, treating, or managing drug or alcohol addiction, or the symptoms thereof, which comprises administering to a mammal, preferably a human, in need of such treatment a therapeutically effective amount of S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer.
  • the invention further encompasses a method of alleviating, treating, or managing the pain and other symptoms associated with the termination of drug use (i.e., withdrawal conditions) which comprises administering to a mammal, preferably a human, in need of such treatment a therapeutically effective amount of S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer.
  • this invention relates to a composition adapted for the treatment of a mammal, preferably a human, suffering from peripheral neuropathy which comprises an amount of S(+) vigabatrin, or a pharmaceutically acceptable salt or solvate thereof, substantially free of its R(-) stereoisomer, said amount being sufficient to alleviate the symptoms of peripheral neuropathy.
  • This invention further encompasses a pharmaceutical composition employing S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer, said composition being formulated for controlled-release of the active ingredient(s) therein.
  • the composition is adapted for the prevention, treatment, or management of peripheral neuropathy, drug addiction, and alleviation of withdrawal symptoms.
  • This controlled-release formulation is particularly useful within the methods described herein.
  • the invention encompasses single unit dosage forms of optically pure S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer, which comprises from about 0.01 g to 1 g of active ingredient in a tablet, capsule, or other single unit dosage form suitable for oral administration.
  • the single unit dosage forms of the invention may also be controlled-release forms as described below.
  • substantially optically pure S(+) vigabatrin is effective in the prevention, treatment, mitigation, and/or management of peripheral neuropathy ("PN"), alcohol and drug addiction or the symptoms thereof, or conditions associated with alcohol or drug withdrawal.
  • optically pure S(+) vigabatrin can be used for the treatment, prevention and/or management of these disorders while reducing or avoiding adverse effects, including undesirable side-effects associated with the administration of racemic vigabatrin.
  • the present invention relates to a method of treating peripheral neuropathy which comprises administering to a mammal a therapeutically effective amount of S(+) vigabatrin, or a pharmaceutically acceptable salt or solvate thereof, substantially free of its R(-) stereoisomer.
  • this invention encompasses single unit dosage forms of optically pure S(+) vigabatrin, or a pharmaceutically acceptable salt, solvate, hydrate or clatherate, thereof, substantially free of its R(-) stereoisomer, which comprises from about 0.1 to about 1 g of active ingredient in a compressed tablet.
  • This dosage form is particularly suitable for the treatment of peripheral neuropathies, such as carpal tunnel syndrome and
  • the methods and compositions of this invention include the benefit of reducing or avoiding adverse effects associated with racemic vigabatrin.
  • the invention also allows the concurrent or sequential use of conventional PN therapies, and drugs such as corticosteroids and antibiotics.
  • the invention further allows the concurrent or sequential use of S(+) vigabatrin and anticonvulsants including, but not limited to, felbamate, gabapentin, lamotrigine, oxcarbazepine, tiagabine, topiramate, zonisamide, carbamazepine, phenobarbital, phenytoin, and valproic acid.
  • compositions of the present invention may also be formulated to provide slow or controlled-release of the active ingredient therein using, for example, hydropropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or the like, or a combination thereof.
  • Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein may be readily selected for use with the S(+) vigabatrin compositions of the invention.
  • single unit dosage forms suitable for oral administration such as tablets, capsules, gelcaps, and the like that are adapted for controlled-release, are encompassed by the present invention.
  • controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled-release formulations may include: 1) extended activity of the drug; 2) reduced dosage frequency; and 3) increased patient compliance.
  • controlled-release formulations are designed to initially release an amount of drug that promptly produces the desired therapeutic effect, and gradual and continual release of other amounts of drug to maintain this level of therapeutic effect over an extended period of time.
  • the drug In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body.
  • controlled-release of the active ingredient may be stimulated by various inducers, for example pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • controlled-release component in the context of the present invention is defined herein as a compound or compounds, including polymers, polymer matrices, gels, permeable membranes, liposomes, microspheres, or the like, or a combination thereof, that facilitates the controlled-release of the active ingredient (e.g., S(+) vigabatrin) in the pharmaceutical composition.
  • the present invention further encompasses the use of S(+) vigabatrin in the prevention and treatment of the symptoms associated with withdrawal from the use of central nervous system ("CNS") stimulants and depressants, such as alcohol and sedatives, and other drugs that act on the CNS which are not considered pure stimulants or depressants, such as barbiturates, psychedelic agents, and marijuana.
  • CNS central nervous system
  • One embodiment of the present invention relates to the administration of a therapeutically effective amount of S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer to a mammal for the treatment of such withdrawal symptoms.
  • the invention also encompasses the use of S(+) vigabatrin to prevent, treat, or manage the development of physical dependence on drugs and alcohol.
  • a therapeutically effective amount of S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer is administered to a patient for the treatment of one or more such dependencies.
  • the present invention further encompasses the use of S(+) vigabatrin in combination with standard detoxification methods to prevent or treat drug withdrawal symptoms in a mammal.
  • S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer is administered before, during, and/or after the detoxification methods.
  • Symptoms of drug withdrawal include, but are not limited to, depression; pain; fever; restlessness; lacrimation; rhinorrhea; uncontrollable yawning; perspiration; piloerection; restless sleep; mydriasis; twitching and muscle spasms; severe aches in the back, abdomen and legs; abdominal and muscle cramps; hot and cold flashes; insomnia; nausea, vomiting, diarrhea; coryza and severe sneezing; and increases in body temperature, blood pressure, respiratory rate, and heart rate (referred to hereinafter, either singly or in combination, as “withdrawal conditions", “withdrawal symptoms”, “conditions associated with drug withdrawal”, or “withdrawal”).
  • the present invention further encompasses the use of S(+) vigabatrin in combination with standard detoxification methods to prevent or treat alcohol withdrawal symptoms in a mammal.
  • S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer is administered before, during, and/or after the detoxification methods.
  • Heavy consumers of alcohol acquire a tolerance and a state of dependence to alcohol.
  • Alcohol withdrawal is not as severe as drug withdrawal, but includes many of the same symptoms.
  • Symptoms of alcohol withdrawal include, but are not limited to, drug craving, tremors, irritability, nausea, sleep disturbance, tachycardia, hypertension, sweating, perceptual distortion, and seizures.
  • Delirium tremens is characterized by severe agitation, confusion, visual hallucinations, fever, profuse sweating, tachycardia, nausea, diarrhea, and dilated pupils.
  • the present invention also encompasses the use of S(+) vigabatrin in combination with other pharmacologically active compounds, such as antidepressants, preferably tricyclic antidepressants.
  • antidepressant compounds include, but are not limited to, tricyclic antidepressants, such as amitriptyline, clomipramine, doxepin, imipramine, trimiprimane, amoxapine, desipramine, maprotiline, nortriptyline, and protryptiline; serotonin-reupake inhibitors, such as fluoxetine, fluvoxamine, paroxetine, sertraline, and venlafaxine; atypical antidepressants, such as bupropion, nefazodone, and trazodone; and other monoamine oxidase inhibitors, such as phenelzine, tranylcypromine, and selgiline, or an isomer of any of the above compounds, either singly or
  • the terms "adverse effects” and “adverse side effects” include, but are not limited to, hyperactivity, agitation, drowsiness, somnolence, fatigue, dizziness, irritability, sedation, weight gain, increased appetite, insomnia, nausea and vomiting, ataxia, decreased appetite, headache, vertigo, and psychosis.
  • salts refer to salts prepared from pharmaceutically acceptable nontoxic acids or bases, including inorganic acids and bases; organic acids and bases; solvates; hydrates; and clatherates thereof. Since the compound of the present invention has both acidic and basic moieties, salts may be prepared from pharmaceutically acceptable nontoxic acids or bases.
  • suitable inorganic bases include metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium, and zinc.
  • Appropriate organic bases may be selected, for example, from N,N-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumaine (N-methylglucamine), lysine and procaine.
  • S(+) vigabatrin is administered as the free acid or free base.
  • racemic means a mixture of the S(+) and R(-) stereoisomers of a compound wherein the S(+) and R(-) stereoisomers are present in approximately a 1 : 1 ratio.
  • substantially optically pure means that the composition contains greater than about 90% of the desired stereoisomer by weight, preferably greater than about 95% of the desired stereoisomer by weight, and most preferably greater than about 99% of the desired stereoisomer by weight, said percent based upon the total weight of vigabatrin.
  • substantially free means less than about 10 weight percent, preferably less than about 5 weight percent, and more preferably less than about 1 weight percent of the undesired stereoisomer.
  • peripheral neuropathy is defined as a disorder characterized by sensory loss, muscle weakness and atrophy, decreased deep tendon reflexes, vasomotor symptoms or any combinations thereof resulting from damage or disease of at least one nerve.
  • the peripheral neuropathy includes carpal tunnel syndrome, Guillain-Barre syndrome, and related disorders.
  • therapeutically effective amount of S(+) vigabatrin means that amount of optically pure S(+) vigabatrin, or a pharmaceutically acceptable salt thereof, substantially free of its R(-) stereoisomer, which provides a therapeutic benefit in the treatment, management, or prevention of a peripheral neuropathy, alcohol addiction, drug addiction, alcohol withdrawal, drug withdrawal, or related disorders.
  • Substantially pure S(+) vigabatrin may be obtained from a racemic mixture of vigabatrin, the chemical synthesis of which can be performed by the methods described in U.S. Patent Nos. 3,960,927 and 5,380,936, the disclosures of which are incorporated herein by express reference thereto.
  • the S(+) stereoisomer of vigabatrin may be obtained from its racemic mixture by chiral separation, such as by gas chromatography, column chromatography, or HPLC.
  • the S(+) stereoisomer of vigabatrin may also be obtained from its racemic mixture by resolution of the stereoisomers using conventional means such as from an optically active resolving acid.
  • S(+) vigabatrin can be synthesized by stereospecific synthesis using methodology well known to those skilled in the art. Chiral synthesis can result in products of high enantiomeric purity. However, in some cases, the enantiomeric purity of the product is not sufficiently high.
  • the skilled artisan will appreciate that the separation methods described above can be used to further enhance the enantiomeric purity of S(+) vigabatrin obtained by chiral synthesis.
  • the magnitude of a prophylactic or therapeutic dose of S(+) vigabatrin in the acute or chronic management of peripheral neuropathy, drug addiction, alcohol addiction, drug withdrawal, and alcohol withdrawal, as described herein, will vary with the severity of the condition to be treated, and the route of administration.
  • the dose, and perhaps the dose frequency, will also vary according to the age, body weight, and response of the individual patient. Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors.
  • the total daily dose range for S(+) vigabatrin, for the conditions described herein is from about 0.01 g to about 4.5 g, in single or divided doses.
  • a daily dose range should be between about 0.05 g to about 4.0 g, in single or divided doses, while most preferably, a daily dose range should be between about 0.1 g to about 3.0 g, in single or divided doses.
  • the maximum daily dosage for children is preferably about 150 mg/kg. It is preferred that the doses for both adults and children be administered from one to four times daily.
  • the therapy may be initiated at a lower dose, e.g., preferably about 0.1 g to about 1 g per day, and increased by about 0.01 g per day up to the recommended daily dose or higher depending on the patient's global response.
  • a starting daily dose of no more than about 40 mg/kg is recommended. This may be increased at appropriate intervals up to the daily recommended dose. It is further recommended that patients over 65 years initially receive low doses, and that their dose be titrated based on individual response(s) and blood level(s). It may be necessary to use dosages outside these ranges in some cases as will be apparent to those skilled in the art.
  • any suitable route of administration may be employed for providing the patient with an effective dosage of S(+) vigabatrin.
  • oral, rectal, nasal, parenteral (subcutaneous, intramuscular, intravenous), sublingual, buccal, mucosal, transdermal, and like forms of administration may be employed.
  • Dosage forms include tablets, troches, lozenges, dispersions, suspensions, suppositories, solutions, capsules, soft elastic gelatin capsules, patches, and the like.
  • orally administered compositions of S(+) vigabatrin may be prepared in a controlled-release formulation.
  • compositions for use in the present invention comprise optically pure S(+) vigabatrin as the active ingredient, or a pharmaceutically acceptable salt or solvate thereof, and may also contain a pharmaceutically acceptable carrier, and optionally, other therapeutic ingredients.
  • compositions of the present invention can be prepared in the form of suspensions, solutions and elixirs; aerosols; and can include carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like, in the case of oral solid preparations (such as powders, capsules, and tablets) with the oral solid preparations being preferred over the oral liquid preparations.
  • a preferred oral solid preparation is capsules.
  • the most preferred oral solid preparation is tablets.
  • tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are employed.
  • tablets may be coated by standard aqueous or non-aqueous techniques, and may be formulated for controlled-release using techniques well known in the art.
  • Preferred controlled-release formulations are capsules and tablets.
  • compositions of the present invention suitable for oral administration may be presented as discrete pharmaceutical unit dosage forms, such as capsules, cachets, soft elastic gelatin capsules, tablets, or aerosols sprays, each containing a predetermined amount of the active ingredient, as a powder or granules, or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion, or a water- in-oil liquid emulsion.
  • Such compositions may be prepared by any of the methods of pharmacy, but all methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more necessary ingredients.
  • the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
  • a tablet may be prepared by compression or molding, optionally, with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as powder or granules, optionally mixed with one or more of a binder, filler, lubricant, inert diluent, or surface active or dispersing agent.
  • Molded tablets may be made by molding, in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent and optionally one or more of a binder, filler, lubricant, inert diluent, or surface active or dispersing agent.
  • Optically pure S(+) vigabatrin for use in the present invention may be formulated as a pharmaceutical composition in a soft elastic gelatin capsule unit dosage form by using conventional methods well known in the art. See, e.g., Ebert, Pharm. Tech, l(5):44-50 (1977).
  • Soft elastic gelatin capsules have a soft, globular gelating shell somewhat thicker than that of hard gelatin capsules, wherein a gelatin is plasticized by the addition of plasticizing agent, e.g., glycerin, sorbitol, or a similar polyol.
  • plasticizing agent e.g., glycerin, sorbitol, or a similar polyol.
  • the hardness of the capsule shell may be changed by varying the type of gelatin used and the amounts of plasticizer and water.
  • the soft gelatin shells may contain a preservative, such as methyl- and propylparabens and sorbic acid, to prevent the growth of fungi.
  • the active ingredient may be dissolved or suspended in a liquid vehicle or carrier, such as vegetable or mineral oils, glycols such as polyethylene glycol and propylene glycol, triglycerides, surfactants such as polysorbates, or a combination thereof.
  • each unit dosage form such as a tablet, capsule, cachet, or soft elastic gelatin capsule, contains from about 0.01 g to about 1 g of the active ingredient, and preferably from about 0.025 g to about 0.9 g of the active ingredient, and more preferably about 0.5 g to about 0.8 g of the active ingredient.
  • Optically pure S(+) vigabatrin for use in the present invention may also be formulated for parenteral administration by injection (subcutaneous, intramuscular, or intravenous), and may be dispensed in a unit dosage form, such as a multidose container or an ampule.
  • Compositions of S(+) vigabatrin for parenteral administration may be in the form of suspensions, solutions, emulsions, or the like in aqueous or oily vehicles, and in addition to the active ingredient may contain one or more formulary agents, such as dispersing agents, suspending agents, stabilizing agents, preservatives, and the like.
  • the compounds of the present invention may also be administered by controlled-release means and/or delivery devices such as those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; and 5,733,566 the disclosures of which are incorporated herein by express reference thereto.
  • the controlled-release formulations of the present invention are capable of releasing the S(+) vigabatrin at the required rate to maintain substantially constant pharmacological activity for a period of time sufficient to provide therapeutic benefits.
  • the S(+) vigabatrin may be prepared in a variety of controlled-release formulations, including, but not limited to, controlled-release pharmaceutical compositions in liquid dosage forms (U.S. Patent No. 5,674,533); controlled-release of active agents by the use of a gastro- resistant tablet (U.S. Patent No. 5,059,595); a liquid reservoir transdermal patch (U.S. Patent No. 5,591 ,767); a controlled-release drug delivery device comprised of swellable polymers (U.S. Patent No.
  • the controlled-release component is biodegradable, induced by exposure to the aqueous environment, pH, temperature, or enzymes in the body.
  • the controlled-release component may swell and form porous openings large enough to release S(+) vigabatrin after administration to a patient.
  • controlled-release is achieved by the use of coated beads or granules.
  • a solution of S(+) vigabatrin in a non-aqueous solvent, such as a mixture of acetone and alcohol, is coated onto granules, such as small inert seeds or beads, made of a combination of sugar and starch.
  • granules such as small inert seeds or beads, made of a combination of sugar and starch.
  • These granules may be coated with a lipid material, such as beeswax, or a cellulosic material, such as ethylcellulose.
  • the variation in the thickness of the coats and the type of material used in the coating is reflected by the speed the bodily fluids are capable of penetrating the coating and dissolving the S(+) vigabatrin.
  • various coating thicknesses of drug granules are present to produce a sustained and controlled-release of S(+) vigabatrin.
  • a portion of the drug granules is coated only with S(+) vigabatrin, while the remaining portion of the drug granules is coated with S(+) vigabatrin and the lipid material.
  • the drug granules without the lipid material coating provide an initial dose of the drug, while the drug granules coated with the lipid material provide a controlled-release of S(+) vigabatrin.
  • microencapsulation of S(+) vigabatrin is used in controlled-release applications.
  • Microscopic size particles can form thin coatings of "wall" material around S(+) vigabatrin.
  • Wall-forming materials include, but are not limited to, gelatin and synthetic polymers, such as polyvinyl alcohol, ethylcellulose, polyvinyl chloride, or a mixture thereof.
  • an additive such as acacia, is used for its ability to concentrate wall-forming material into tiny liquid droplets that form a film or coat around the drug or drugs to be encapsulated.
  • sol-gels may be used, wherein S(+) vigabatrin is incorporated into a sol-gel matrix that is a solid at room temperature.
  • This matrix is implanted into a patient, preferably a mammal, and more preferably a human, having a body temperature high enough to induce gel formation of the sol-gel matrix, thereby releasing S(+) vigabatrin into the patient.
  • the drug may be embedded in an inert plastic matrix.
  • S(+) vigabatrin is granulated with an inert plastic material, such as polyethylene, polyvinyl acetate, polymethacrylate, or a mixture thereof, and the granulation is compressed into tablets. S(+) vigabatrin is slowly released from the inert plastic matrix by leaching via bodily fluids.
  • repeat action formulations release two full doses of S(+) vigabatrin sequentially.
  • This method utilizes specialized tablets that release an initial dose of S(+) vigabatrin from the tablet shell, and a second dose is released from an inner core of the tablet, separated from the outer shell by a slowly permeable baffler coating.
  • any suitable method known to those of ordinary skill in the art for formulating controlled-release dosage forms may be employed according to the invention.
  • the relative potency and specificity of optically pure S(+)vigabatrin, as compared to R(-)vigabatrin and racemic vigabatrin, as a potent irreversible inhibitor of GABA-T may be determined by pharmacological study. As a consequence of this enzyme's inhibition activity, the compounds will produce proportional increases in concentration of the inhibitory neurotransmitter GABA.
  • the relative specificity of the compounds may be determined against cerebral neuronal and glial cell GABA-T.
  • GABA-T Because transamination by GABA-T otherwise represents the significant catabolic pathway of cerebral GABA, the irreversible inhibition of GABA-T by S(+) vigabatrin results in an elevation of brain GABA levels throughout the brain. However, glial GABA-T is less sensitive to S(+) vigabatrin, presumably for the lack of an uptake system for the drug into glial cells. Thus the drug may preferentially influence amounts of cerebral neuronal, synaptically-released GABA. Rogawski, M.A., et al, Pharm. Rev., 42(3):223-286 (1990); Jung, M.T, et al, J. Neurochem.
  • test systems are based on tissue culture techniques with two distinct cerebral cell types: a) cerebral neurons with characteristics of GABAergic neurons derived from cultured cerebral cortex of 15 day mouse embryos, and b) astrocytes obtained by culture of cortical tissue of newborn mice.
  • a) cerebral neurons with characteristics of GABAergic neurons derived from cultured cerebral cortex of 15 day mouse embryos and b) astrocytes obtained by culture of cortical tissue of newborn mice.
  • Racemic vigabatrin and its stereoisomers may be incubated with the tissue cultures of 10 minutes at 37°C, or alternatively the compounds may be present throughout the full incubation period with the cell cultures.
  • S(+)vigabatrin, R(-)vigabatrin and racemic vigabatrin may be compared for their ability to inhibit both preparations of neuronal GABA-T and glial cell GABA-T.
  • the drugs may be compared at the concentrations that inhibit by 50% the enzyme activity of the appropriate GABA-T (IC 50 ). Further, the concentrations of the drugs may be compared for their ability to increase both the cell content, and the neurotransmitter, GABA. (See aforementioned references, and Gram, L., et al, Brit. J. Clin., Pharm. 27, (Suppl. 1):13S-18S (1989)).
  • the active ingredient, S(+) vigabatrin is sieved through a suitable sieve and blended with the lactose until a uniform blend is formed. Suitable volumes of water are added and the powders are granulated. After drying, the granules are then screened and blended with the remaining excipients. The resulting granules are then compressed into tablets of desired shape. Tablets of other strengths may be prepared by altering the ratio of active ingredient to the excipient(s) or the compression weight.
  • a suspension of S(+) vigabatrin and HPMC may be applied onto 18/20 mesh Nupariel beads in a fluid bed dryer with a Wurster insert at an inlet temperature of 60°C.
  • An Opadry ® Lavender YS-1-4729 HMPC Base filmcoating suspension is then applied after drug loading as a protective coat at a 5% weight gain.
  • the S(+) vigabatrin beads are then overcoated with a retardant coating mixture of Eudragit ® RS 30D and Eudragit ® RL 30D at a ratio of 90:10, RS to RL, at a 5% weight gain level.
  • a retardant coating mixture of Eudragit ® RS 30D and Eudragit ® RL 30D at a ratio of 90:10, RS to RL, at a 5% weight gain level.
  • Eudragit ® RS 30D and Eudragit ® RL 30D along with talc (included as an antitacking agent) and triethyl citrate (plasticizer) is done at an inlet temperature of 35°C in a Wurster insert.
  • the resulting S(+) vigabatrin beads are given a final overcoat of Opadry ® Lavender YS 1-4729 at a 5% weight gain level, and cured on paper- lined trays in a 45°C dry oven for 2 days. After the curing process is complete, the S(+) vigabatrin beads are filled into gelatin capsules at 0.5 g S(+) vigabatrin strength.
  • All Opadry ® products used in this example and the following examples are available from Colorcon, West Point, PA.
  • All Eudragit ® products used in this example and the following examples are available from Rohm Pharma. The final formula is provided in Table 2 below:
  • S(+) vigabatrin beads are prepared by dissolving S(+) vigabatrin in water, adding Opadry ® Y-5-1442, light pink and mixing for about 1 hour to obtain a 20% w/w suspension. This suspension is sprayed onto Nupareil 18/20 mesh beads using a Wurster insert.
  • the loaded S(+) vigabatrin beads are overcoated to form a protective coating with a 5% w/w gain of Opadry ® Light Pink using a Wurster insert.
  • a retardant coating is then applied to the S(+) vigabatrin beads by coating them with a 5% weight gain of a mixture of Eudragit ® RS 30D and Eudragit ® RL 30D at a ratio of 90:10, RS to RL.
  • the addition of triethyl citrate (plasticizer) and talc (anti-tacking agent) is also included in the Eudragit suspension.
  • a Wurster insert is used to apply the coating suspension.
  • the S(+) vigabatrin beads are given a final overcoat of Opadry ® Light Pink to a 5% weight gain using a Wurster insert, and cured in a45°C oven for two days. The cured beads are then filled into gelatin capsules at a 0.05 g S(+) vigabatrin strength.
  • Table 3 The complete formula is shown in Table 3 below:
  • a mixture of 80% (w/w) S(+) vigabatrin, 10% (w/w) polyvinylpyrrolidone (PVP K30), and 10% (w/w) 450-mesh lactose may be wetted with 500 mL water and mixed in a mixer-granulator for 10 minutes.
  • a 5% (w/w) solution of polyethylene glycol (PEG 6000) is added as an aqueous solution to the stirred mixture at a rate of 25 mL/min using a 0.8 mm nozzle at a pressure of 2 bars.
  • the mixer speed is 175 rpm and the speed of the mill is 3000 rpm.
  • the kneading and rounding step is done by maintaining mixer and mill speeds at a constant rate for 15 minutes.
  • the resulting microgranulate is dried in an artificial ventilation incubator and sieved through a 225- mesh/cm 2 screen until a microgranulate with a granule size distribution ranging from 90 ⁇ m and a spheroidal shape was obtained.
  • EXAMPLE 6 Coating of Microgranulates
  • Two kilograms of the microgranulate prepared in accordance with Example 5 are sti ⁇ ed for 1 minute in a Glatt GPCG3 (Glad, GmbH, Buizen Lorrach, Germany) fluidizer into which air heated to a temperature of 40°C to 45°C was blown at a rate of 40 mVhour.
  • the granulate was sprayed at a pressure of 2 bars and a rate of 10 to 13 g/min with 400 mL of a solution having the following weight percent composition:
  • a microgranulate, prepared as described in Example 5 and coated with three successive layers as described in Example 6, may be added to a final proportion of 12.5% (w/w), to a mixture containing 6.2% (w/w) microcrystalline cellulose, 0.8% (w/w) sodium carboxymethylcellulose, 0.5% (w/w) sodium citrate, 0.8% (w/w) citric acid, 0.2 % (w/w) methylparaben.

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Neurology (AREA)
  • Addiction (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)

Abstract

La présente invention concerne des procédés et compositions pour la prévention, le traitement et/ou la gestion des symptômes de neuropathie périphérique et d'affections connexes, de narcomanie ou d'alcoolisme ou leurs symptômes, ou des symptômes associés au sevrage alcoolique ou au sevrage de drogue, utilisant du vigabatrin S(+) optiquement pur, ou un sel de celui-ci pharmaceutiquement acceptable.
EP99945177A 1998-08-25 1999-08-24 Procedes et compositions employant le vigabatrin s(+) optiquement pur Withdrawn EP1107748A2 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US9778698P 1998-08-25 1998-08-25
US97786P 1998-08-25
US11445698P 1998-12-30 1998-12-30
US114456P 1998-12-30
PCT/US1999/019346 WO2000010554A2 (fr) 1998-08-25 1999-08-24 Procedes et compositions employant le vigabatrin s(+) optiquement pur

Publications (1)

Publication Number Publication Date
EP1107748A2 true EP1107748A2 (fr) 2001-06-20

Family

ID=26793638

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99945177A Withdrawn EP1107748A2 (fr) 1998-08-25 1999-08-24 Procedes et compositions employant le vigabatrin s(+) optiquement pur

Country Status (5)

Country Link
EP (1) EP1107748A2 (fr)
JP (1) JP2003520189A (fr)
AU (1) AU5784499A (fr)
CA (1) CA2341400A1 (fr)
WO (1) WO2000010554A2 (fr)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007136845A2 (fr) 2006-05-19 2007-11-29 Somaxon Pharmaceuticals, Inc. Doxépine à faible dose utilisée pour le traitement de troubles du sommeil chez des patients âgés
US20100179214A1 (en) 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin trans isomers and isomeric mixtures and methods of using the same to treat sleep disorders
CA2687118A1 (fr) 2006-05-19 2007-12-13 Somaxon Pharmaceuticals, Inc. Procedes d'utilisation de doxepine faiblement dosee pour ameliorer le sommeil
US20100179215A1 (en) 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin isomers and isomeric mixtures and methods of using the same to treat sleep disorders
WO2008011150A1 (fr) 2006-07-20 2008-01-24 Somaxon Pharmaceuticals, Inc. Méthodes destinées à améliorer la pharmacocinétique de la doxépine
WO2008052139A2 (fr) 2006-10-25 2008-05-02 Somaxon Pharmaceuticals, Inc. Doxepine à dose ultrafaible, et ses procédés d'utilisation pour traiter des troubles du sommeil
US20110077200A1 (en) 2006-12-06 2011-03-31 Somaxon Pharmaceuticals, Inc. Combination therapy using low-dose doxepin for the improvement of sleep
EP2148659A2 (fr) 2007-04-13 2010-02-03 Somaxon Pharmaceuticals, Inc. Formulations de doxépine à faible dose et procédé de préparation et d'utilisation de celle-ci
CA3016851A1 (fr) * 2016-03-10 2017-09-14 Orphelia Pharma Formes galeniques solides de vigabatrine
CN110869003A (zh) * 2019-01-28 2020-03-06 上海奥科达生物医药科技有限公司 一种不含防腐剂的固体剂型氨己烯酸药物组合物

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2133002B (en) * 1982-12-30 1986-01-29 Merrell Toraude & Co Process for preparing 4-amino-5-hexenoic acid
US4595697A (en) * 1984-03-28 1986-06-17 Merrell Dow Pharmaceuticals Inc. Treatment of seizure disorders and pharmaceutical compositions useful therein
EP0509180A1 (fr) * 1991-04-18 1992-10-21 Merrell Dow Pharmaceuticals Inc. Utilisation d'inhibiteurs de la GABA-T pour le traitement de la schizophrenie et l'intoxication de la phencyclidine
US6541520B1 (en) * 1998-08-05 2003-04-01 Brookhaven Science Associates Treatment of addiction and addiction-related behavior

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0010554A2 *

Also Published As

Publication number Publication date
WO2000010554A2 (fr) 2000-03-02
AU5784499A (en) 2000-03-14
WO2000010554A3 (fr) 2000-11-30
JP2003520189A (ja) 2003-07-02
CA2341400A1 (fr) 2000-03-02

Similar Documents

Publication Publication Date Title
CN104958282B (zh) 他喷他多组合物
JPH083035A (ja) 医薬の応答反応の増強
US20080031950A1 (en) Novel anelgesic combination
JP2007526277A (ja) 経口投与を目的としたトルペリゾンの徐放性医薬組成物
EA014707B1 (ru) Новая форма введения рацекадотрила
AU758181B2 (en) Therapeutic agents
CN1141937C (zh) 含有曲马朵的药物组合物用于制备治疗偏头痛或类偏头痛的药物
KR100894465B1 (ko) 레보도파의 지속적 효과를 위한 조성물 및 제형
WO2000010554A2 (fr) Procedes et compositions employant le vigabatrin s(+) optiquement pur
US6376552B1 (en) Treatment of gallstones
WO2000056318A1 (fr) Traitement de la douleur neuropathique ou de la cellulomyalgie
JP2003521469A (ja) 睡眠時無呼吸症を治療する方法
CN114432266B (zh) 一种稳定的盐酸环苯扎林缓释胶囊
AU773490B2 (en) Treatment of osteoarthritis
WO2000056311A1 (fr) Regulation du metabolisme
JPH06509073A (ja) 光学的に純粋なs(−)オンダンセトロンを使用する嘔吐、吐き気および他の障害の治療のための方法および組成物
JP2002539254A (ja) 血小板粘着性を低下させるための治療
JP2002539250A (ja) 肺高血圧症の治療
GB2141930A (en) Novel pharmaceutical preparations with anti-tumour effect
CN112915083A (zh) 药物组合物、成套药盒及其应用
JP2002539255A (ja) 体重増加に関係する癌の治療
JP2002539252A (ja) 胆石の治療
JP2002539249A (ja) 裂孔ヘルニアの治療
WO2000056308A1 (fr) Prevention de maladies cardio-vasculaires
MXPA00006201A (en) Pharmaceutical composition containing sibutramine and orlistat

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: 20010228

AK Designated contracting states

Kind code of ref document: A2

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

AX Request for extension of the european patent

Free format text: AL PAYMENT 20010228;LT PAYMENT 20010228;LV PAYMENT 20010228;MK PAYMENT 20010228;RO PAYMENT 20010228;SI PAYMENT 20010228

17Q First examination report despatched

Effective date: 20020222

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: 20020705