WO1996023492A1 - Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin - Google Patents

Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin Download PDF

Info

Publication number
WO1996023492A1
WO1996023492A1 PCT/US1996/001053 US9601053W WO9623492A1 WO 1996023492 A1 WO1996023492 A1 WO 1996023492A1 US 9601053 W US9601053 W US 9601053W WO 9623492 A1 WO9623492 A1 WO 9623492A1
Authority
WO
WIPO (PCT)
Prior art keywords
oxybutynin
desethyloxybutynin
pharmaceutically acceptable
acceptable salt
ethyl
Prior art date
Application number
PCT/US1996/001053
Other languages
French (fr)
Inventor
Gunnar Aberg
John R. Mc Cullough
Yue Fang
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
Priority claimed from US08/381,542 external-priority patent/US5532278A/en
Priority to AU49664/96A priority Critical patent/AU706741B2/en
Priority to DE69610290T priority patent/DE69610290T2/en
Priority to EP96906202A priority patent/EP0806948B1/en
Priority to SK1035-97A priority patent/SK283313B6/en
Priority to CA002211400A priority patent/CA2211400C/en
Priority to JP8523636A priority patent/JPH11511730A/en
Priority to BR9607001A priority patent/BR9607001A/en
Application filed by Sepracor, Inc. filed Critical Sepracor, Inc.
Priority to UA97084420A priority patent/UA45387C2/en
Priority to AT96906202T priority patent/ATE196252T1/en
Priority to CZ19972421A priority patent/CZ290093B6/en
Priority to DK96906202T priority patent/DK0806948T3/en
Priority to PL96321498A priority patent/PL182768B1/en
Priority to NZ303372A priority patent/NZ303372A/en
Publication of WO1996023492A1 publication Critical patent/WO1996023492A1/en
Priority to FI973163A priority patent/FI973163A/en
Priority to NO19973516A priority patent/NO315927B1/en
Priority to MXPA/A/1997/005820A priority patent/MXPA97005820A/en
Priority to HK98111210A priority patent/HK1017986A1/en
Priority to GR20000402596T priority patent/GR3034974T3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder

Definitions

  • the invention relates to a method for treating urinary incontinence using optically pure (S)- oxybutynin and (S)-desethyloxybutynin (S-DEO) , to pharmaceutical compositions containing optically pure (S)-oxybutynin or S-DEO, and to a process for preparing single enantiomers of DEO.
  • Racemic oxybutynin is used therapeutically in the treatment of intestinal hypermotility and in the treatment of urinary incontinence due to detrusor instability. Racemic oxybutynin exerts a direct antispasmodic effect on smooth muscle and inhibits the action of acetylcholine on smooth muscle. It exhibits only one-fifth of the anticholinergic activity of atropine on the rabbit detrusor muscle, but four to ten times the antispasmodic activity. It is quite selective for muscarinic receptors in the presence of nicotinic receptors and as a result, no blocking effects are observed at skeletal neuromuscular junctions or autonomic ganglia.
  • Racemic oxybutynin relaxes bladder smooth muscle and, in patients with conditions characterized by involuntary bladder contractions, cystometric studies have demonstrated that racemic oxybutynin increases vesicle capacity, diminishes the frequency of involuntary contractions of the detrusor muscle, and delays the initial desire to void. It is therefore useful in the treatment and prevention of both incontinency and frequent voluntary urination.
  • the efficacy of racemic oxybutynin in the bladder has been attributed to a combination of antimuscarinic, direct spasmolytic and local anesthetic effects on the detrusor smooth muscle.
  • Optically pure (S)-oxybutynin (S-OXY) and (S)- desethyloxybutynin (S-DEO) provide this treatment while substantially reducing the adverse effects that primarily arise from anticholinergic activity and that are associated with the administration of racemic oxybutynin. These include, but are not limited to, xerostomia, mydriasis, drowsiness, nausea, constipation, palpitations and tachycardia.
  • the amelioration of cardiovascular side effects of racemic oxybutynin, such as tachycardia and palpitations, by the administration of (S)-oxybutynin or S-DEO is of particular therapeutic value.
  • the active compounds of these compositions and methods are optical isomers of oxybutynin and desethyloxybutynin.
  • the preparation of racemic oxybutynin is described in British Patent Specification 940,540.
  • the active compounds are (1) the S enantiomer of 4- (diethylamino)-2-butynyl ⁇ -cyclohexyl- ⁇ - hydroxybenzeneacetate also known as 4-(diethylamino)- 2-butynyl phenylcyclohexylglycolate, and hereinafter referred to as oxybutynin; and (2) the S enantiomer of 4-(ethylamino)-2-butynyl ⁇ -cyclohexyl- ⁇ - hydroxybenzeneacetate, and hereinafter referred to as desethyloxybutynin.
  • (S)-Desethyloxybutynin has not been previously described; its synthesis is carried out according to the method described below.
  • the invention relates to a method for treating urinary incontinence while avoiding concomitant liability of adverse effects, which comprises administering to a human in need of such treatment a therapeutically effective amount of (S)- oxybutynin, (S)-desethyloxybutynin or a pharmaceutically acceptable salt of either, substantially free of the corresponding R enantiomer.
  • compositions contain at least 90% by weight of (S)-oxybutynin or (S)- desethyloxybutynin and 10% by weight or less of (R)- oxybutynin or (R)-desethyloxybutynin.
  • compositions contain at least 99% by weight of the S enantiomer and 1% or less of the R enantiomer.
  • the substantially optically pure (S)-oxybutynin or (S)- desethyloxybutynin may be administered parentally, rectally, intravesically, transdermally, orally or by aerosol, orally and transdermally being preferred, at a rate of about l mg to about 100 mg per day.
  • the invention in another aspect, relates to a pharmaceutical unit dosage form in the form of a tablet or capsule comprising a therapeutically effective amount of (S)-oxybutynin, (S)- desethyloxybutynin or a pharmaceutically acceptable salt of either, substantially free of the corresponding R stereoisomer, and a pharmaceutically acceptable carrier.
  • the tablet or capsule preferably contains from 0.5 to 25 mg of (S)-oxybutynin or (S)- desethyloxybutynin and is prepared by conventional methods, well-known in the art.
  • the invention also relates to a dosage form in the form of a transdermal device. The transdermal administration is improved by the inclusion of a permeation enhancer in the transdermal delivery device, for example as described in PCT application WO 92/20377.
  • the invention relates to a process for preparing desethyloxybutynin, preferably a single enantiomer of DEO, most preferably S-DEO, comprising the steps of, first, reacting methyl o- cyclohexyl- ⁇ -hydroxybenzeneacetate with 4-[N-ethyl- (4-methoxyphenyl)methylamino]-2-butyn-l-ol in the presence of an anhydrous base to produce 4-[N-ethyl- (4-methoxyphenyl)methylamino]-2-butynyl ⁇ -cyclohexyl- ⁇ -hydroxybenzeneacetate; and then sequentially with a carbonochloridate and methanol to produce 4- (ethylamino)-2-butynyl ⁇ -cyclohexyl- ⁇ - hydroxybenzeneacetate (desethyloxybutynin) .
  • the process may additionally comprise reacting N-ethyl-4- methoxybenzenemethanamine with 2-propyn-l-ol and formaldehyde or a formaldehyde equivalent in the presence of a copper(I) salt to produce the 4-[N- ethyl-(4-methoxyphenyl)methylamino]-2-butyn-l-ol needed for the first step.
  • the S enantiomers of oxybutynin and DEO may be obtained by resolution of the intermediate mandelic acid followed by esterification.
  • the esterification can be carried out as described by Kachur et al. (op. cit.) for OXY or by the improved method depicted in Scheme A below for S-DEO.
  • the overall process for DEO involves:
  • racemic DEO from racemic methyl ⁇ -cyclohexyl- ⁇ - hydroxybenzeneacetate
  • Paraformaldehyde is used as a convenient source of formaldehyde, but it can be replaced by any source of formaldehyde, as is well known in the art.
  • ⁇ -chloroethyl carbonochloridate is used for the dealkylation, but other carbonochloridates (e.g.vinyl) could be employed.
  • the S enantiomers of OXY and DEO may be obtained by the resolution of racemic oxybutynin or DEO using conventional means such as fractional crystallization of diastereomeric salts with chiral acids.
  • Other standard methods of resolution known to those skilled in the art, including, but not limited to, simple crystallization and chromatography on a chiral substrate can also be used.
  • the magnitude of a prophylactic or therapeutic dose of (S)-oxybutynin or S-DEO in the acute or chronic management of disease will vary with the severity and nature 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.
  • the total daily dose range for (S)-oxybutynin or S-DEO for the conditions described herein is from about 1 mg to about 100 mg in single or divided doses, preferably in divided doses.
  • the therapy should be initiated at a lower dose, perhaps at about 0.25 mg to about 25 mg, and increased up to about 100 mg depending on the patient's global response.
  • any suitable route of administration may be employed for providing the patient with an effective dosage of (S)-oxybutynin or S-DEO.
  • oral, rectal, parenteral (subcutaneous, intramuscular, intravenous) , transdermal, aerosol and like forms of administration may be employed.
  • the drug may be administered directly into the bladder through the urethra, as described for racemic oxybutynin by Massad et al. [J_j_ Urol. 148. 595-597 (1992)].
  • Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, transdermal delivery systems, and the like.
  • compositions of the present invention comprise (S)-oxybutynin or S-DEO as the active ingredient, or a pharmaceutically acceptable salt thereof, and may also contain a pharmaceutically acceptable carrier, and optionally, other therapeutic ingredients.
  • pharmaceutically acceptable salts or "a pharmaceutically acceptable salt thereof” refer to salts prepared from pharmaceutically acceptable non- toxic acids.
  • suitable pharmaceutically acceptable acid addition salts for the compound of the present invention include acetic, benzenesulfonic (besylate) , benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic, and the like.
  • the hydrochloride has particular utility and was, in fact, the salt used in the studies described below.
  • compositions of the present invention include suspensions, solutions, elixirs, or solid dosage forms.
  • Carriers such as starches, sugars, and macrocrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like are suitable in the case of oral solid preparations (such as powders, capsules, and tablets) , and oral solid preparations are preferred over the oral liquid preparations.
  • tablets and capsules represent one of the more advantageous oral dosage unit forms, in which case solid pharmaceutical carriers are employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques.
  • the compounds of the present invention may also be administered by controlled release means and delivery devices such as those described in U.S.Patent Nos. : 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, and PCT application WO 92/20377, the disclosures of which are hereby incorporated by reference.
  • compositions of the present invention suitable for oral administration may be presented as discrete unit dosage forms such as capsules, cachets, or tablets, 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, just as is known for the racemic mixture.
  • 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 a binder, lubricant, inert diluent, surface active agent 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. All of the foregoing techniques are well known to persons of skill in the pharmaceutical art.
  • Each tablet may contain from about 0.5 mg to about 25 mg of the active ingredient.
  • the (S)-oxybutynin or desethyloxybutynin is blended with the lactose and cellulose until a uniform blend is formed.
  • the lake is added and further blended.
  • the calcium stearate is blended in, and the resulting mixture is compressed into tablets using a 9/32 inch (7 mm) shallow concave punch. Tablets of other strengths may be prepared by altering the ratio of active ingredient to the excipients or to the final weight of the tablet.
  • the compounds were tested on each receptor at 10 concentrations in duplicate to obtain competition curves.
  • the reference compound for the receptor under investigation was simultaneously tested at 8 concentrations in duplicate to obtain a competition curve in order to validate this experiment.
  • the specific radioligand binding of each receptor was defined as the difference between total binding and nonspecific binding determined in the presence of an excess of unlabelled ligand. IC 50 values
  • Binding assays were performed using the following methods:
  • the experiment conditions were:
  • the compounds were tested in duplicate on each receptor at a concentration of 10 "5 M.
  • the reference compound for the receptor under investigation was simultaneously tested at 8 concentrations in duplicate to obtain a competition curve in order to validate this experiment.
  • the specific radioligand binding of each receptor was defined as the difference between total binding and nonspecific binding determined in the presence of an excess of unlabelled ligand. Mean values, expressed as a percentage of inhibition of specific binding, are presented in Table 5. IC 50 values (concentration required to inhibit 50% of specific binding) were determined by non linear regression analysis of their competition curves. These parameters were obtained by curve fitting using SigmaplotTM software.
  • the major metabolite of racemic oxybutynin is RS-desethyl oxybutynin (DEO) .
  • DEO RS-desethyl oxybutynin
  • the R and S enantiomers of DEO have not been described, and the antispasmodic and calcium entry blocking activities of the individual enantiomers, R- and S-DEO, were, prior to our studies, unknown.
  • each enantiomer of the metabolite retains the relative pharmacological profile of its "parent" oxybutynin enantiomer.
  • the ethanol controls were used when, because of poor solubility, stock solutions of test substances had to be prepared in ethanol, as a result of which the tissue baths experienced an effective concentration of 17 mM ethanol. Finally, the responses to increasing concentrations of carbachol followed by exposure to 137.7 mM KCl were recorded a second time.
  • the peak tension developed by each strip during the second set of determinations was expressed as a percent of the peak tension developed during the first concentration-effect determination. Then, for each antagonist the resultant data were analyzed for treatment-related differences by one-way analysis of variance (ANOVA) . Since only one concentration of antagonist was studied in each strip of bladder, the procedures of Arunlakshana and Schild (1959) were used in modified form to estimate the pA2 and slope of the Schild regression. First, the concentrations of agonist producing a half-maximal response (the EC JQ ) was estimated for each strip from the second set of concentration-effect data.
  • ANOVA analysis of variance
  • the EC 50 was obtained from linear regression lines fit to the logarithm of the concentration of drug and the responses bracketing the half maximum level of response.
  • a "concentration ratio” (CR) was calculated as the ratio of the EC 50 of the treated tissue divided by the EC 50 of the untreated tissue.
  • log (CR-1) the logarithm of this ratio minus one [i.e., log (CR-1) ] was plotted against the logarithm of the concentration of antagonist to which the strip had been exposed to produce "Schild plots”.
  • a regression analysis relating log(CR-1) to the logarithm of the concentration of the antagonist was employed to estimate the pA2 and the slope of the regression line.
  • racemic precursor to DEO (2.05 g, 43% yield) was obtained as an oil from racemic IV (2.98 g, 12.0 mmol) and V (2.48 g, 10.6 mmol).
  • (+)-DEO- HC1(0.63 g) was obtained as an off-white solid from ( ⁇ ) precursor (2.28 g, 5.08 mmol).
  • S-Oxybutynin may be prepared by the same route, substituting 4-(diethylamino)-2-butyn-l-ol for the protected intermediate V.

Abstract

A method for treating urinary incontinence while avoiding concomitant liability of adverse effects associated with racemic oxybutynin is disclosed. The method comprises administering a therapeutically effective amount of (S)-oxybutynin, (S)-desethyloxybutynin or a pharmaceutically acceptable salt thereof, substantially free of the corresponding R enantiomer. Pharmaceutical compositions in the form of tablets and transdermal devices comprising (S)-oxybutynin or (S)-desethyloxybutynin and an acceptable carrier are also disclosed, as is a synthesis of desethyloxybutynin.

Description

TREATING URINARY INCONTINENCE USING (S)-OXYBUTYNIN AND (S)-DESETHY OXYBUTYNIN
FIELD OF THE INVENTION
The invention relates to a method for treating urinary incontinence using optically pure (S)- oxybutynin and (S)-desethyloxybutynin (S-DEO) , to pharmaceutical compositions containing optically pure (S)-oxybutynin or S-DEO, and to a process for preparing single enantiomers of DEO.
BACKGROUND OF THE INVENTION
Racemic oxybutynin is used therapeutically in the treatment of intestinal hypermotility and in the treatment of urinary incontinence due to detrusor instability. Racemic oxybutynin exerts a direct antispasmodic effect on smooth muscle and inhibits the action of acetylcholine on smooth muscle. It exhibits only one-fifth of the anticholinergic activity of atropine on the rabbit detrusor muscle, but four to ten times the antispasmodic activity. It is quite selective for muscarinic receptors in the presence of nicotinic receptors and as a result, no blocking effects are observed at skeletal neuromuscular junctions or autonomic ganglia.
Racemic oxybutynin relaxes bladder smooth muscle and, in patients with conditions characterized by involuntary bladder contractions, cystometric studies have demonstrated that racemic oxybutynin increases vesicle capacity, diminishes the frequency of involuntary contractions of the detrusor muscle, and delays the initial desire to void. It is therefore useful in the treatment and prevention of both incontinency and frequent voluntary urination. The efficacy of racemic oxybutynin in the bladder has been attributed to a combination of antimuscarinic, direct spasmolytic and local anesthetic effects on the detrusor smooth muscle. Because of the antimuscarinic activity of the racemic drug, xerostomia (dry mouth) and mydriasis (dilated pupils) , which involve muscarinic cholinergic receptors, are very common side effects. In fact, at least one researcher has referred to the "inevitable symptoms of mydriasis, xerostomia, tachycardia, etc." that accompany the administration of racemic oxybutynin [Lish et al. Arch. Int. Pharmacodyn. 156. 467-488 (1965), 481]. The high incidence of anticholinergic side effects (40 to 80%) often results in dosage reduction or discontinuation of therapy.
Pharmacological studies of the individual enantiomers have suggested that the R-enantiomer is the efficacious enantiomer. Noronha-Blob et al. fJ. Pharmacol. Exp. Ther. 256. 562-567 (1991)] concluded that the cholinergic antagonism of racemic oxybutynin (measured in vitro by its affinity for M,, M2 and M3 receptors subtypes and in vivo for diverse physiological responses) could be attributed mainly to the activity of the R-enantiomer. For all responses they found the rank order of potency of racemic oxybutynin and its enantiomers to be the same, namely, (R)-oxybutynin greater than or equal to racemic oxybutynin, which was much greater than (S)- oxybutynin, with (S)-oxybutynin being 1 to 2 orders of magnitude less potent than (R)-oxybutynin. SU MARY OF THE INVENTION
It has now been unexpectedly found that the substantially optically pure S enantiomer of oxybutynin and of its desethyl metabolite provide a superior therapy for the treatment of urinary incontinence.
Optically pure (S)-oxybutynin (S-OXY) and (S)- desethyloxybutynin (S-DEO) provide this treatment while substantially reducing the adverse effects that primarily arise from anticholinergic activity and that are associated with the administration of racemic oxybutynin. These include, but are not limited to, xerostomia, mydriasis, drowsiness, nausea, constipation, palpitations and tachycardia. The amelioration of cardiovascular side effects of racemic oxybutynin, such as tachycardia and palpitations, by the administration of (S)-oxybutynin or S-DEO is of particular therapeutic value.
The active compounds of these compositions and methods are optical isomers of oxybutynin and desethyloxybutynin. The preparation of racemic oxybutynin is described in British Patent Specification 940,540. Chemically, the active compounds are (1) the S enantiomer of 4- (diethylamino)-2-butynyl α-cyclohexyl-α- hydroxybenzeneacetate also known as 4-(diethylamino)- 2-butynyl phenylcyclohexylglycolate, and hereinafter referred to as oxybutynin; and (2) the S enantiomer of 4-(ethylamino)-2-butynyl α-cyclohexyl-α- hydroxybenzeneacetate, and hereinafter referred to as desethyloxybutynin. The generic name given to the hydrochloride salt of racemic oxybutynin by the USAN Council is oxybutynin chloride; it is sold under the trade name of Ditropan®. The isomer of oxybutynin having the S absolute stereochemistry (Registry Number 119618-22-3) is dextrorotatory, and is shown in Formula I:
Figure imgf000006_0001
The S enantiomer of desethyloxybutynin is shown in Formula II:
Figure imgf000006_0002
II
The synthesis of (S)-oxybutynin has been described [Kachur et al. J^. Pharmacol. EXP. Ther. 247. 867-872 (1988)], but (S)-oxybutynin itself is not presently commercially available. All of the clinical results that have been reported have been obtained with the racemic mixture, although the pharmacology of the individual enantiomers has been described in guinea pigs and rats [see Kachur et al. J. Pharmacol. Exp. Ther. 247. 867-872 (1988) and Noronha-Blob et al. ιL_ Pharmacol. EXP. Ther. 256. 562-567 (1991)]. (S)-Desethyloxybutynin has not been previously described; its synthesis is carried out according to the method described below. In one aspect the invention relates to a method for treating urinary incontinence while avoiding concomitant liability of adverse effects, which comprises administering to a human in need of such treatment a therapeutically effective amount of (S)- oxybutynin, (S)-desethyloxybutynin or a pharmaceutically acceptable salt of either, substantially free of the corresponding R enantiomer. The term "substantially free of its R enantiomer" as used herein means that the compositions contain at least 90% by weight of (S)-oxybutynin or (S)- desethyloxybutynin and 10% by weight or less of (R)- oxybutynin or (R)-desethyloxybutynin.
In a more preferred embodiment, the compositions contain at least 99% by weight of the S enantiomer and 1% or less of the R enantiomer. The substantially optically pure (S)-oxybutynin or (S)- desethyloxybutynin may be administered parentally, rectally, intravesically, transdermally, orally or by aerosol, orally and transdermally being preferred, at a rate of about l mg to about 100 mg per day.
In another aspect, the invention relates to a pharmaceutical unit dosage form in the form of a tablet or capsule comprising a therapeutically effective amount of (S)-oxybutynin, (S)- desethyloxybutynin or a pharmaceutically acceptable salt of either, substantially free of the corresponding R stereoisomer, and a pharmaceutically acceptable carrier. The tablet or capsule preferably contains from 0.5 to 25 mg of (S)-oxybutynin or (S)- desethyloxybutynin and is prepared by conventional methods, well-known in the art. The invention also relates to a dosage form in the form of a transdermal device. The transdermal administration is improved by the inclusion of a permeation enhancer in the transdermal delivery device, for example as described in PCT application WO 92/20377.
In a further aspect the invention relates to a process for preparing desethyloxybutynin, preferably a single enantiomer of DEO, most preferably S-DEO, comprising the steps of, first, reacting methyl o- cyclohexyl-α-hydroxybenzeneacetate with 4-[N-ethyl- (4-methoxyphenyl)methylamino]-2-butyn-l-ol in the presence of an anhydrous base to produce 4-[N-ethyl- (4-methoxyphenyl)methylamino]-2-butynyl α-cyclohexyl- α-hydroxybenzeneacetate; and then sequentially with a carbonochloridate and methanol to produce 4- (ethylamino)-2-butynyl α-cyclohexyl-α- hydroxybenzeneacetate (desethyloxybutynin) . The process may additionally comprise reacting N-ethyl-4- methoxybenzenemethanamine with 2-propyn-l-ol and formaldehyde or a formaldehyde equivalent in the presence of a copper(I) salt to produce the 4-[N- ethyl-(4-methoxyphenyl)methylamino]-2-butyn-l-ol needed for the first step.
DETAILED DESCRIPTION OF THE INVENTION
The S enantiomers of oxybutynin and DEO may be obtained by resolution of the intermediate mandelic acid followed by esterification. The esterification can be carried out as described by Kachur et al. (op. cit.) for OXY or by the improved method depicted in Scheme A below for S-DEO. Scheme A
Figure imgf000009_0001
Figure imgf000009_0002
ac eto ne
Figure imgf000009_0003
Figure imgf000009_0004
V I I
The graphic representations of racemic, ambiscalemic and scalemic or enantiomerically pure compounds used herein are taken from Maehr J_j_ Chem. Ed. 62. 114-120 (1985) . Thus, solid and broken wedges (such as shown in formula I) are used to denote the absolute configuration of a chiral element; wedge outlines and dotted or broken lines (such as shown in formula III) denote enantiomerically pure compounds of indeterminate absolute configuration.
The overall process for DEO involves:
(a) reacting N-ethyl-4- methoxybenzenemethanamine with 2-propyn-l-ol and paraformaldehyde in an inert solvent in the presence of cuprous chloride to produce 4-[N-ethyl-(4- methoxyphenyl)methylamino]-2-butyn-l-ol (V) ;
(b) reacting a single enantiomer of methyl α- cyclohexyl-α-hydroxybenzeneacetate (IV) with 4-[N- ethyl-(4-methoxyphenyl)methylamino]-2-butyn-l-ol (V) in the presence of a catalytic amount of sodium methoxide in toluene to produce a single enantiomer of 4-[N-ethy1-(4-methoxypheny1)methylamino]-2-butyny1 α-cyclohexyl-α-hydroxybenzeneacetate (VI) ; and
(c) reacting 4-[N-ethyl-(4- methoxyphenyl)methylamino]-2-butyny1 α-cyclohexyl-α- hydroxybenzeneacetate (VI) sequentially with α- chloroethyl carbonochloridate in dichloroethane, followed by methanol to produce a single enantiomer of DEO (VII) .
The process is obviously applicable to producing racemic DEO from racemic methyl α-cyclohexyl-α- hydroxybenzeneacetate as well. Paraformaldehyde is used as a convenient source of formaldehyde, but it can be replaced by any source of formaldehyde, as is well known in the art. Similarly α-chloroethyl carbonochloridate is used for the dealkylation, but other carbonochloridates (e.g.vinyl) could be employed.
Alternatively, the S enantiomers of OXY and DEO may be obtained by the resolution of racemic oxybutynin or DEO using conventional means such as fractional crystallization of diastereomeric salts with chiral acids. Other standard methods of resolution known to those skilled in the art, including, but not limited to, simple crystallization and chromatography on a chiral substrate can also be used.
The magnitude of a prophylactic or therapeutic dose of (S)-oxybutynin or S-DEO in the acute or chronic management of disease will vary with the severity and nature 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. In general, the total daily dose range for (S)-oxybutynin or S-DEO for the conditions described herein is from about 1 mg to about 100 mg in single or divided doses, preferably in divided doses. In managing the patient, the therapy should be initiated at a lower dose, perhaps at about 0.25 mg to about 25 mg, and increased up to about 100 mg depending on the patient's global response. It is further recommended that patients over 65 years and those with impaired renal or hepatic function initially receive low doses and that they 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. Further, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with individual patient response. The terms "a therapeutically effective amount" and "an amount sufficient to treat incontinence but insufficient to cause adverse effects" are encompassed by the above-described dosage amounts and dose frequency schedule.
Any suitable route of administration may be employed for providing the patient with an effective dosage of (S)-oxybutynin or S-DEO. For example, oral, rectal, parenteral (subcutaneous, intramuscular, intravenous) , transdermal, aerosol and like forms of administration may be employed. Additionally, the drug may be administered directly into the bladder through the urethra, as described for racemic oxybutynin by Massad et al. [J_j_ Urol. 148. 595-597 (1992)]. Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, transdermal delivery systems, and the like.
The pharmaceutical compositions of the present invention comprise (S)-oxybutynin or S-DEO as the active ingredient, or a pharmaceutically acceptable salt thereof, and may also contain a pharmaceutically acceptable carrier, and optionally, other therapeutic ingredients.
The terms "pharmaceutically acceptable salts" or "a pharmaceutically acceptable salt thereof" refer to salts prepared from pharmaceutically acceptable non- toxic acids. Suitable pharmaceutically acceptable acid addition salts for the compound of the present invention include acetic, benzenesulfonic (besylate) , benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic, and the like. The hydrochloride has particular utility and was, in fact, the salt used in the studies described below.
The compositions of the present invention include suspensions, solutions, elixirs, or solid dosage forms. Carriers such as starches, sugars, and macrocrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like are suitable in the case of oral solid preparations (such as powders, capsules, and tablets) , and oral solid preparations are preferred over the oral liquid preparations.
Because of their ease of administration, tablets and capsules represent one of the more advantageous oral dosage unit forms, in which case solid pharmaceutical carriers are employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques.
In addition to the common dosage forms set out above, the compounds of the present invention may also be administered by controlled release means and delivery devices such as those described in U.S.Patent Nos. : 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, and PCT application WO 92/20377, the disclosures of which are hereby incorporated by reference.
Pharmaceutical compositions of the present invention suitable for oral administration may be presented as discrete unit dosage forms such as capsules, cachets, or tablets, 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. In general, 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, just as is known for the racemic mixture.
For example, 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 a binder, lubricant, inert diluent, surface active agent 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. All of the foregoing techniques are well known to persons of skill in the pharmaceutical art. Each tablet may contain from about 0.5 mg to about 25 mg of the active ingredient. EXAMPLES
Example 1 ORAL UNIT DOSAGE FORMULATION Tablets:
Ingredients per tablet per batch of
10,000 tablets
(S)-Oxybutynin or 5mg 50g (S)-Desethyloxybutynin
Microcrystalline cellulose 30mg 300g
Lactose 70mg 700g
Calcium stearate 2mg 20g
FD&C Blue #1 Lake 0.03mg 300mg
The (S)-oxybutynin or desethyloxybutynin is blended with the lactose and cellulose until a uniform blend is formed. The lake is added and further blended. Finally, the calcium stearate is blended in, and the resulting mixture is compressed into tablets using a 9/32 inch (7 mm) shallow concave punch. Tablets of other strengths may be prepared by altering the ratio of active ingredient to the excipients or to the final weight of the tablet.
The surprising utility of the S enantiomer of both OXY and DEO has been established by the following studies. ENANTIOMERS OF OXYBUTYNIN
Binding of (R)- and fS)-Oxybutynin to Human Mt. M7. M3 and M,. Muscarinic Receptor Subtypes
MATERIALS AND METHODS
The experiments were carried out on membranes prepared from SF9 cells infected with baculovirus to express the human recombinant M,, M2, M3 and M4 muscarinic receptor subtypes.
Binding Assays
Table 1
Figure imgf000016_0001
Following incubation, the assays were rapidly filtered under vacuum through GF/B glass fiber filters (Whatman) and washed with an ice-cold buffer using a Brandel Cell Harvester. Bound radioactivity was determined with a liquid scintillation counter (LS 6000, Beckman) using a liquid scintillation cocktail (Formula 99, DuPont NEN) .
Experimental Protocol
The compounds were tested on each receptor at 10 concentrations in duplicate to obtain competition curves. In each experiment, the reference compound for the receptor under investigation was simultaneously tested at 8 concentrations in duplicate to obtain a competition curve in order to validate this experiment.
Analysis and expression of results
The specific radioligand binding of each receptor was defined as the difference between total binding and nonspecific binding determined in the presence of an excess of unlabelled ligand. IC50 values
(concentrations required to inhibit 50% of specific binding) were determined by non linear regression analysis of the competition curves. These parameters were obtained by curve fitting using Sigmaplot™ software. IC50 for R- and S-OXY are given in Table 2.
Table 2
Binding of R-oxybutynin and S-oxybutynin to human muscarinic subtypes Ml - M4
R-OXY S-OXY Re . Compound
Receptor IC50(nM) ICjo(nM) IC50(nM)
Ml 0.99 47.6 Pirenzepine 11.9
M2 9.9 178 Methoctramine 14.6
M3 1.8 149 4-DAMP 1.6
M4 1.2 100 4-DAMP 0.87
These results indicate that S-OXY has less affinity for muscarinic receptor subtypes than does R-OXY. Binding of (R)- and fS.-Oxybutynin to Calcium
Channels
MATERIALS AND METHODS
Binding assays
Binding assays were performed using the following methods:
Table 3
Reference
Receptors Membranes Compounds References
Ca channel rat cerebral cortex diltiazem Schoemaker and
(T + L, diltiazem site) Langer (1985)
Ca channel rat cerebral cortex D600 Reynolds et a I
(T + L, verapamil site) (1986)
The experiment conditions were:
Table 4
Receptors Ligands Concentrations Nonspecific Incubation
Ca channel
(T + L, diltiazem site) l3Hl diltiazem 5nM dilitiazem 120 min (10 M) 25°C
Ca channel [3H]D 888 0.5 nM D 600 60 min
(T + L,verapamil site) (10/ M) 22°C
Following incubation, the assays were rapidly filtered under vacuum through GF/B or GF/C glass fiber filters (Whatman) and washed with an ice-cold buffer using a Brandel Cell Harvester. Bound radio¬ activity was determined with a liquid scintillation counter (LS6000, Beckman) using a liquid scintillation cocktail (Formula 989, DuPont NEN) .
Experimental Protocols
The compounds were tested in duplicate on each receptor at a concentration of 10"5M. In each experiment, the reference compound for the receptor under investigation was simultaneously tested at 8 concentrations in duplicate to obtain a competition curve in order to validate this experiment.
Analysis and expression of results
The specific radioligand binding of each receptor was defined as the difference between total binding and nonspecific binding determined in the presence of an excess of unlabelled ligand. Mean values, expressed as a percentage of inhibition of specific binding, are presented in Table 5. IC50 values (concentration required to inhibit 50% of specific binding) were determined by non linear regression analysis of their competition curves. These parameters were obtained by curve fitting using Sigmaplot™ software.
Table 5. Binding of R-oxybutynin and S-oxybutynin to calcium channels [Inhibition (in %) of diltiazem and verapamil binding to calcium channel receptors.]
R-OXY S-OXY Ref. Compound
Receptor (lO^M) (10"SM) ICJ0(nM)
Calcium (diltiazem) 86 59 diltiazem 55.8
Calcium (verapamil) 86 68 D600 36.4
These results indicate that S-OXY has calcium entry blocking activity similar to R-OXY.
ENANTIOMERS OF DESETHYLOXYBUTYNIN
The major metabolite of racemic oxybutynin is RS-desethyl oxybutynin (DEO) . The R and S enantiomers of DEO have not been described, and the antispasmodic and calcium entry blocking activities of the individual enantiomers, R- and S-DEO, were, prior to our studies, unknown. We have synthesized these enantiomers and have studied their antimuscarinic, spasmolytic and calcium entry blocking effects in models of receptor binding and bladder function. We have found that each enantiomer of the metabolite retains the relative pharmacological profile of its "parent" oxybutynin enantiomer.
Binding at Muscarinic Receptor Subtypes
The percent inhibition of specific radioligand binding induced by three concentrations of each compound (R-, S-, and RS-DEO) was tested at cloned human muscarinic receptor subtypes (M1-M4) , as described above for the enantiomers of oxybutynin. The tables below (Tables 6 and 7) give the percent inhibition at each subtype. In addition, ICS0 values were determined for M, and M2 human receptor subtypes and are presented in Table 6.
Table 6
M,„ Mm
10βM 107M 10SM ICM 10βM 107M 105M ιcM (nM) (nM)
R-DEO 63 100 100 1.2 21 97 102 14.7
S-DEO — 82 101 25.4 - 36 101 177
RS-DEO 43 100 100 1.8 - 94 99 7.0 Table 7
Figure imgf000021_0001
These results indicate that S-DEO has less affinity for muscarinic receptor subtypes than either R- or racemic DEO.
Binding at Calcium Channels
The percent inhibition of specific radioligand binding induced by each compound (R-, S-, and RS-DEO) was tested at the diltiazem and verapamil sites of the L-type calcium channel. The results are shown in Table 8.
Table 8
Receptor R-Deo S-DEO RS-DEO lO^M lO ^M 10,5M
Calcium (diltiazem) 86 72 88
Calcium (verapamil) 96 76 89
These results indicate that S-DEO has calcium entry blocking activity similar to that of R- and racemic DEO.
Functional Characterization of Antimuscarinic/Antispasmodic Activity
The effects of R-, S- and RS-Oxybutynin (OXY) and of R-, S-, and RS-DEO were studied in an in vitro model of bladder function. As described below, isolated strips of guinea pig bladder smooth muscle were mounted in a tissue bath and contracted either with the muscarinic agonist carbachol or with increasing concentrations of external potassium.
MATERIALS AND METHODS
Bladder strips. Experiments were performed using methods similar to those described by Kachur et al, 1988 and Noronha-Blob and Kachur, 1991. Strips of tissue (approximately 10 mm long and 1.5 mm wide) were removed from the body of the urinary bladder of male Hartley guinea pigs weighing 400-600 g. (Elm Hill Breeding Laboratories, Chelmsford, MA) . The tissues were suspended in an oxygenated buffer of the following composition, in mM: NaCl, 133; KC1, 4.7; CaCl2, 2.5; MgS04, 0.6; NaH2P04, 1.3; NaHC03, 16.3; and glucose, 7.7. They were maintained at 37.5° C. Contractions were recorded with isometric transducers (Model FT-10) and an ink-writing polygraph (Model 7) (Astro-Med, Inc., Grass Instrument Div. , West Warwick, RI) . A resting tension of 0.5 grams was maintained on all tissues at all times.
In each experiment up to seven strips were removed from a single bladder, suspended in individual tissue chambers and allowed to equilibrate with the bathing solution for one hour before proceeding with the experiment.
Carbachol-induced contractions. One series of experiments focused on the anticholinergic actions of oxybutynin. In these experiments, in order to assess the viability of each tissue and to serve as a frame of reference, contractions of each strip of tissue were recorded initially in response to exposure to tissue medium in which the NaCl was replaced by KCl to yield a concentration of 137.7 mM KCl in the medium. This was followed by return to the standard medium, and then by exposures to progressively increasing concentrations of carbachol, with separate exposures to each concentration only until the peak response had been recorded. Then, leaving one strip untreated and/or one strip exposed to 17 mM ethanol to serve as control tissue(s) , the remaining strips each were exposed for one hour to one concentration of an antagonist. The ethanol controls were used when, because of poor solubility, stock solutions of test substances had to be prepared in ethanol, as a result of which the tissue baths experienced an effective concentration of 17 mM ethanol. Finally, the responses to increasing concentrations of carbachol followed by exposure to 137.7 mM KCl were recorded a second time.
Potassium-induced contractions. A second series of experiments focused on the spasmolytic action of the substances being studied. Contractions were recorded in response to sequentially increasing the concentration of potassium in the medium.
Data analysis. To determine whether antagonists decreased the peak response to agonists, the peak tension developed by each strip during the second set of determinations was expressed as a percent of the peak tension developed during the first concentration-effect determination. Then, for each antagonist the resultant data were analyzed for treatment-related differences by one-way analysis of variance (ANOVA) . Since only one concentration of antagonist was studied in each strip of bladder, the procedures of Arunlakshana and Schild (1959) were used in modified form to estimate the pA2 and slope of the Schild regression. First, the concentrations of agonist producing a half-maximal response (the ECJQ) was estimated for each strip from the second set of concentration-effect data. The EC50 was obtained from linear regression lines fit to the logarithm of the concentration of drug and the responses bracketing the half maximum level of response. For each drug-treated strip, a "concentration ratio" (CR) was calculated as the ratio of the EC50 of the treated tissue divided by the EC50 of the untreated tissue. For each experiment where two or more strips were exposed to the same chemical but at different concentrations, the logarithm of this ratio minus one [i.e., log (CR-1) ] was plotted against the logarithm of the concentration of antagonist to which the strip had been exposed to produce "Schild plots". A regression analysis relating log(CR-1) to the logarithm of the concentration of the antagonist was employed to estimate the pA2 and the slope of the regression line. Finally, experiments were grouped by chemical and the mean + S.E. of the pA2 and slope were calculated. The 95% confidence limits (CL) for the slope were estimated from its S.E. using standard methods. For experiments in which only one strip was exposed to a given chemical, a pKD was calculated as (concentration of antagonist)/(CR-1) and the negative logarithm of the KD was then pooled with the pA2 values to yield an expanded set of pA2 values.
Results
The effects of racemic oxybutynin and DEO and their respective enantiomers on carbachol-induced contraction are summarized in Table 9 below. Values given are the summary of Schild analyses which gives pA2 values [mean ± SE] and slope [mean + SE] .
Table 9
Antagonist No. of expts. pA2 Slope
R-OXY 4 8.80 +. 0.27 1.28 . 0.26
S-OXY 4 7.09 i 0.13 1.13 t 0.17
RS-OXY 5 8.81 .+ 0.29 1.34 jh. 0.15
R-DEO 4 9.04 +_ 0.32 1.16 ± 0.1 1
S-DEO 4 7.31 i θ.35 0.87 ^ 0.1 1
RS-DEO 4 8.55 _+ 0.32 1.35 ± 0.25
These results indicate that both S-OXY and S-DEO are less potent antagonists of bladder muscarinic receptors than are R- and racemic OXY and R- and racemic DEO.
The effects of racemic oxybutynin and its enantiomers on potassium-induced contraction are summarized in Table 10 below. (Values given are the magnitude of contraction induced by 137.7 mM K+ after 60 min exposure to compound divided by the magnitude of contraction induced before exposure to drug. )
Table 10
Mean % pretreatment
Antagonist ± SE (n«3)
R-OXY 32 ± 8*
S-OXY 26 ± 9*
RS-OXY 20 ± 1*
R-DEO 36 ± 5*
S-DEO 42 ± 5*
RS-DEO 47 ± 8*
♦Significantly different from corresponding value for untreated tissues (p<0.01)
These results indicate that oxybutynin and its enantiomers and desethyl oxybutynin and its enantiomers are equipotent as bladder smooth muscle spasmolytics.
CONCLUSIONS
While it is well known that the normal emptying of the bladder is mediated through cholinergic mechanisms, the bladder instability that is seen in patients suffering from incontinence appears to be related to non-cholinergic contractions of the bladder. Andersson et al. [Neurourol Urodyn 5_, 579- 586 (1986) ] have shown in animals that the atropine- resistant detrusor muscle is highly sensitive to calcium antagonists.
The study of the receptor binding affinities of (R)- and (S)-oxybutynin to the receptor sites for the calcium channel blockers diltiazem and verapamil described above allows one to conclude that S- oxybutynin and (S)-desethyloxybutynin have therapeutic effects on involuntary micturition, while (unlike the R-isomers and the racemates) having very little effect on the normal voiding mechanism. Both also exhibit significantly decreased anticholinergic side effects as compared with the corresponding R- isomer and racemate. The avoidance of cardiovascular side effects that arise from the anticholinergic action of racemic oxybutynin is of particular note. We conclude that S-oxybutynin and S-desethyl oxybutynin are effective medicaments for the treatment of urinary incontinence in humans with greatly reduced side effects over the racemates or the pure R-enantiomers.
Methyl (R)-α-cyclohexyl-α-hydroxybenzeneacetate (IV)
To a mixture of (R)-α-cyclohexyl-α- hydroxybenzeneacetic acid (III) (12.2 g, 52.1 mmol) and K2C03 (10.8 g, 78.2 mmol) in 100 mL of acetone was added methyl iodide (Mel) (13.0 L, 208 mmol) dropwise at 0° C (ice bath) . After the addition (ca. Ih) of Mel, the reaction mixture was stirred at room temperature overnight. The mixture was filtered through a pad of Celite and rinsed with acetone twice. The filtrate was concentrated to give a white slurry which was diluted with water and extracted with heptane. The combined extracts were washed with water, brine, dried and concentrated to give the product (R)-IV (11.9 g, 92% yield) as a white solid.
Methyl (S)-o-cyclohexyl-o-hydroxybenzeneacetate (IV):
Following the same procedure as above, (S)-IV (11.2 g, 100% yield) was obtained as a white solid from (S)-III (10.6 g, 45.3 mmol). 4-[N-Ethyl-( -methoxypheny1) ethylamino]-2-butynyl (R)-α-cyclohexyl-α-hydroxybenzeneacetate (VI) :
To a solution of (R)-IV(11.9 g, 47.7 mmol) and 4-[N- ethyl-(4-methoxypheny1) ethylamino]-2-butyn-l-ol (V) (9.30 g, 39.9 mmol) in 120 mL of toluene was added
NaOMe (0.222 g, 4.11 mmol). The reaction mixture was stirred at reflux for 5 h and a total of 6 mL of the solvent was removed by Dean-Stark apparatus. The reaction mixture was cooled to room temperature, diluted with ethyl acetate, washed with water, brine, dried and concentrated. The residue was chromatographed on silica gel (elution with 1%, 2.5% and then 5% MeOH in CH2C12) to afford the product (R)- VI(14.1 g, 79% yield) as an oil.
4-[N-Ethyl-(4-methoxyphenyl)methylamino]-2-butynyl (S)- -cyclohexyl-α-hydroxybenzeneacetate (VI) :
Following the same procedure as above, (S)-VI(4.24 g, 58% yield) was obtained as an oil from (S)-IV(4.07 g, 16.4 mmol) and V (4.24 g, 18.2 mmol).
Racemic 4-[N-Ethyl-(4-methoxyphenyl)methylamino]-2- butynyl (S)-α-cyclohexyl-α-hydroxybenzene-acetate:
Following the same procedure as above, the racemic precursor to DEO (2.05 g, 43% yield) was obtained as an oil from racemic IV (2.98 g, 12.0 mmol) and V (2.48 g, 10.6 mmol).
4-(Ethylamino)-2-butynyl (R)-o-cyclohexyl-α- hydroxybenzeneacetate hydrochloride salt (VII)-HC1: A solution of (R)-VI (14.0 g, 31.2 mmol) and α- chloroethyl carbonochloridate (4.0 mL, 37.4 mmol) in 1,2-dichloroethane was stirred at reflux for 1 h.
After cooling, the reaction mixture was concentrated and 200 mL of MeOH was added to the residue. The reaction mixture was stirred at reflux for 20 min and cooled to room temperature. The mixture was concentrated and the residue was chromatographed on silica gel (elution with 1% and then 50% MeOH in CH2C12) and then triturated with ether to afford the product (R)-VII-HCl (8.93 g, 87% yield) as a tan solid. This tan solid was further purified by recrystallization from EtOH/Et20 and by sequential treatment with 10% aqueous K2C03 and EtOAc, activated carbon, and a solution of IN HC1 in ether to give (R)-DEO-HCl (6.44 g) as an off-white solid.
4-(Ethylamino)-2-butyny1 (S)-α-cyclohexyl-α- hydroxybenzeneacetate hydrochloride salt (VII)-HCl:
Following the same procedure as above, (S)-DEO- HCl(5.27g, 57% yield) was obtained as an off-white solid from (S)-VI (11.4g, 25.4 mmol).
Racemic 4-(Ethylamino)-2-butynyl α-cyclohexyl-α- hydroxybenzeneacetate hydrochloride salt:
Following the same procedure as above, (+)-DEO- HC1(0.63 g) was obtained as an off-white solid from (±) precursor (2.28 g, 5.08 mmol).
S-Oxybutynin may be prepared by the same route, substituting 4-(diethylamino)-2-butyn-l-ol for the protected intermediate V.
The 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2- butyn-1-ol (V) used as an intermediate is synthesized as follows:
N-Ethyl-4-methoxybenzenemethanamine:
To a mixture of anisaldehyde (15.6 g, 115 mmol) and ethyla ine (2.0 M in THF, 87 mL, 174 mmol) in 1,2- dichloroethane (450 L) was added glacial acetic acid (10.0 mL, 174 mmol) under nitrogen atmosphere. The reaction mixture was stirred at room temperature for 30 min and then cooled to 0° C with ice bath. NaBH(OAc)3(36.9 g, 174 mmol) was added portionwise and the reaction mixture was stirred at room temperature overnight. The mixture was concentrated and the residue was diluted with a basic solution (10 g NaOH in 100 mL of water) to make the solution slightly basic. This aqueous layer was extracted with ether. The combined extracts were washed with water, brine, dried and concentrated. The residue was chromatographed on silica gel (elution with 5% MeOH in CH2C12 and then 50% MeOH in CH2C12 containing 4% Et3N) to afford the product (11.2 g, 59% yield) as an oil.
4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butyn-l-ol (V):
A mixture of N-ethy1-4-methoxybenzenemethanamine (13.3 g, 80.6 mmol), paraformaldehyde (3.63 g) , propargyl alcohol (6.33 g, 113 mmol) and CuCl (0.311 g) in 350 mL of 1,4-dioxane was stirred at reflux for 30 min. The reaction mixture was cooled to room temperature and concentrated. The residue was diluted with 20C mL of 50% NH40H and extracted with
EtOAc. The combined extracts were washed with water, brine, dried and concentrated. The residue was chromatographed on silica gel (elution with 2.5% MeOH in CH2C12 and then 5% MeOH in CH2C12) to afford the product V (15.1 g, 81% yield) as an oil.

Claims

1. A method for treating urinary incontinence while avoiding concomitant liability of adverse effects, which comprises administering to a human in need of such treatment a therapeutically effective amount of a compound chosen from the group consisting of (S)-oxybutynin and (S)-desethyloxybutynin or a pharmaceutically acceptable salt thereof, substantially free of its R enantiomer.
2. A method according to claim 1 for treating urinary incontinence while avoiding concomitant liability of adverse effects, which comprises administering to a human in need of such treatment a therapeutically effective amount of (S)-oxybutynin or a pharmaceutically acceptable salt thereof, substantially free of its R enantiomer.
3. The method of claim 2 wherein (S)- oxybutynin is administered by inhalation or by parenteral, transdermal, rectal or oral administration.
4. The method of claim 3 wherein the amount of (S)-oxybutynin or a pharmaceutically acceptable salt thereof administered is from about 1 mg to about 100 mg per day.
5. The method according to claim 2 wherein (S)-oxybutynin, or pharmaceutically acceptable salt thereof, is administered orally.
6. The method according to claim 2 wherein (S)-oxybutynin, or pharmaceutically acceptable salt thereof, is administered transdermally.
7. A method according to claim 1 for treating urinary incontinence while avoiding concomitant liability of adverse effects, which comprises administering to a human in need of such treatment a therapeutically effective amount of (S)- desethyloxybutynin or a pharmaceutically acceptable salt thereof, substantially free of its (R) enantiomer.
8. The method of claim 7 wherein (S)- desethyloxybutynin is administered by inhalation or by parenteral, transdermal, rectal or oral administration.
9. The method of claim 8 wherein the amount of (S)-desethyloxybutynin or a pharmaceutically acceptable salt thereof administered is from about l mg to about 100 mg per day.
10. The method according to claim 7 wherein (S)-desethyloxybutynin, or pharmaceutically acceptable salt thereof, is administered orally.
11. The method according to claim 7 wherein (S)-desethyloxybutynin, or pharmaceutically acceptable salt thereof, is administered transderma1ly.
12. A pharmaceutical unit dosage form in the form of a tablet or capsule which comprises a therapeutically effective amount of a compound chosen from the group consisting of (S)-oxybutynin and (S)- desethyloxybutynin, or a pharmaceutically acceptable salt thereof, substantially free of its (R) stereoisomer, and a pharmaceutically acceptable carrier.
13. A pharmaceutical unit dosage form according to claim 12 comprising from 0.5 to 25 mg of (S)- oxybutynin.
14. A pharmaceutical unit dosage form according to claim 12 comprising from 0.5 to 25 mg of (S)- desethyloxybutynin.
15. A pharmaceutical dosage form in the form of a transdermal delivery device which comprises a therapeutically effective amount of a compound chosen from the group consisting of (S)-oxybutynin and (S)- desethyloxybutynin, or a pharmaceutically acceptable salt thereof, substantially free of its (R) stereoisomer, and a pharmaceutically acceptable carrier.
16. A pharmaceutical dosage form according to claim 15 wherein said pharmaceutically acceptable carrier comprises a permeation enhancer.
17. A process for preparing desethyloxybutynin comprising the steps of:
(a) reacting methyl α-cyclohexyl-α- hydroxybenzeneacetate with 4-[N-ethyl-(4- methoxyphenyl)methylamino]-2-butyn-l-ol in the presence of an anhydrous base to produce 4-[N-ethyl- (4-methoxypheny1)methylamino]-2-butynyl α-cyclohexyl- α-hydroxybenzeneacetate; and (b) reacting said 4-[ -ethy1-(4- methoxyphenyl)methylamino]-2-butynyl α-cyclohexyl-α- hydroxybenzeneacetate sequentially with a carbonochloridate and methanol to produce 4- (ethylamino)-2-butynyl α-cyclohexyl-α— hydroxybenzeneacetate.
18. A process according to claim 17 comprising the additional step of reacting N-ethyl-4- methoxybenzenemethanamine with 2-propyn-l-ol and formaldehyde or a formaldehyde equivalent in the presence of a copper(I) salt to produce 4-[N-ethyl- ( -methoxypheny1)methylamino]-2-butyn-1-ol.
19. A process according to claim 18 comprising:
(a) reacting N-ethyl-4- methoxybenzenemethanamine with 2-propyn-l-ol and paraformaldehyde in an inert solvent in the presence of cuprous chloride to produce 4-[N-ethyl-(4- methoxyphenyl)methylamino]-2-butyn-1-ol;
(b) reacting methyl α-cyclohexyl-α- hydroxybenzeneacetate with said 4-[N-ethyl-(4- methoxyphenyl)methylamino]-2-butyn-l-ol in the presence of sodium methoxide in toluene to produce 4- [N-ethyl-(4-methoxyphenyl)methylamino]-2-butynyl α- cyclohexyl-α-hydroxybenzeneacetate; and
(c) reacting said 4-[N-ethyl-(4- methoxypheny1)methylamino]-2-butyny1 α-cyclohexy1-α- hydroxybenzeneacetate sequentially with α-chloroethyl carbonochloridate in dichloroethane, followed by methanol to produce 4-(ethylamino)-2-butynyl α- cyclohexyl-α-hydroxybenzeneacetate (desethyloxybutyin) .
20. A process according to claim 17 wherein said desethyloxybutynin is enriched in a single enantiomer.
21. A process according to claim 20 wherein said desethyloxybutynin is S-desethyloxybutynin.
PCT/US1996/001053 1995-01-31 1996-01-25 Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin WO1996023492A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
UA97084420A UA45387C2 (en) 1995-01-31 1996-01-25 METHOD OF TREATMENT OF URINARY INCONTINENCE Related prevent the likelihood of negative effects, the active ingredients of medicines (option) pharmaceutical dosage form (option) AND METHOD FOR PRODUCING (S) -DEZETYLOKSYBUTYNINU
NZ303372A NZ303372A (en) 1995-01-31 1996-01-25 Use of (s)-oxybutynin and (s)-desethyloxybutynin to treat urinary incontinence
EP96906202A EP0806948B1 (en) 1995-01-31 1996-01-25 Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin
SK1035-97A SK283313B6 (en) 1995-01-31 1996-01-25 Treating urinary incontinence using (S)-oxybutynin and (S)-desethyloxybutynin
CA002211400A CA2211400C (en) 1995-01-31 1996-01-25 Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin
JP8523636A JPH11511730A (en) 1995-01-31 1996-01-25 Treatment of urinary incontinence using (S) -oxybutynin and (S) -desethyloxybutynin
BR9607001A BR9607001A (en) 1995-01-31 1996-01-25 Treatment of urinary incontinence with (o) -oxybutynin and (s) -desetyloxybutynin
CZ19972421A CZ290093B6 (en) 1995-01-31 1996-01-25 Medicament based on (S)-oxybutynin or (S)-desethyloxybutynin for treating urinary incontinence and pharmaceutical preparation
AT96906202T ATE196252T1 (en) 1995-01-31 1996-01-25 TREATMENT OF HORN INCONTINENCE BY USING (S)-OXYBUTYNIN AND (S)-DESETHYLOXYBUTYNIN
AU49664/96A AU706741B2 (en) 1995-01-31 1996-01-25 Treating urinary incontinence using (S)-oxybutynin and (S)-desethyloxybutynin
DE69610290T DE69610290T2 (en) 1995-01-31 1996-01-25 TREATMENT OF HORN INCONTINENCE USING (S) -OXYBUTYNIN AND (S) -DESETHYLOXYBUTYNIN
DK96906202T DK0806948T3 (en) 1995-01-31 1996-01-25 Treatment of urinary incontinence using (S) -oxybutynin and (S) -desethyloxybutynin
PL96321498A PL182768B1 (en) 1995-01-31 1996-01-25 Pharmaceutic agent for use against urine incontinence and method of obtaining (s)-desethyloxybutinine
FI973163A FI973163A (en) 1995-01-31 1997-07-30 Treatment of incontinence using (S) -oxybutynin and (S) -desethyloxybutynin
MXPA/A/1997/005820A MXPA97005820A (en) 1995-01-31 1997-07-30 The use of (s) -oxibutinin and (s) -desetiloxibutinin in the preparation of compositions for the treatment of urinary incontinence, the compositions obtained and the procedure for preparing desetiloxibitin
NO19973516A NO315927B1 (en) 1995-01-31 1997-07-30 Use of (S) -oxybutynin and (S) -desetyloxybutynin for the preparation of a medicament for the treatment of urinary incontinence as well as the pharmaceutical composition containing the same
HK98111210A HK1017986A1 (en) 1995-01-31 1998-10-13 Treating urinary incontinence using (s)-oxybutyninand (s)-desethyloxybutynin.
GR20000402596T GR3034974T3 (en) 1995-01-31 2000-11-24 Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08/381,542 1995-01-31
US08/381,542 US5532278A (en) 1995-01-31 1995-01-31 Methods and compositions for treating urinary incontinence using optically pure (S)-oxybutynin
US08/480,194 1995-06-07
US08/480,194 US5677346A (en) 1995-01-31 1995-06-07 Treating urinary incontinence using (S)-desethyloxybutynin

Publications (1)

Publication Number Publication Date
WO1996023492A1 true WO1996023492A1 (en) 1996-08-08

Family

ID=27009434

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/001053 WO1996023492A1 (en) 1995-01-31 1996-01-25 Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin

Country Status (25)

Country Link
US (1) US5677346A (en)
EP (1) EP0806948B1 (en)
JP (1) JPH11511730A (en)
KR (1) KR100390271B1 (en)
CN (1) CN1151786C (en)
AT (1) ATE196252T1 (en)
AU (1) AU706741B2 (en)
BR (1) BR9607001A (en)
CA (1) CA2211400C (en)
CZ (1) CZ290093B6 (en)
DE (1) DE69610290T2 (en)
DK (1) DK0806948T3 (en)
ES (1) ES2150663T3 (en)
FI (1) FI973163A (en)
GR (1) GR3034974T3 (en)
HK (1) HK1017986A1 (en)
HU (1) HUP9800794A3 (en)
NO (1) NO315927B1 (en)
NZ (1) NZ303372A (en)
PL (1) PL182768B1 (en)
PT (1) PT806948E (en)
RU (1) RU2181589C2 (en)
SK (1) SK283313B6 (en)
UA (1) UA45387C2 (en)
WO (1) WO1996023492A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998000126A1 (en) * 1996-06-28 1998-01-08 Sepracor, Inc. Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin
WO1998011888A1 (en) * 1996-09-19 1998-03-26 American Home Products Corporation Method of treating urinary incontinence
US6013830A (en) * 1998-03-30 2000-01-11 Sepracor Inc. Asymmetric grignard synthesis with cyclic 1,2 aminoalcohols
WO2000071108A2 (en) * 1999-05-20 2000-11-30 Sepracor Inc. Methods for treatment of asthma using s-oxybutynin
DE10103262A1 (en) * 2001-01-25 2002-08-01 Axel Schmidt-Dossi Suppository containing oxybutynin, useful for treatment of incontinence, avoids side effects and allows an increase in dose
WO2002062389A1 (en) * 2001-02-08 2002-08-15 Ono Pharmaceutical Co., Ltd. Remedies for urinary diseases comprising lpa receptor controlling agents
US6521724B2 (en) 2000-03-10 2003-02-18 E. I. Du Pont De Nemours And Company Polymerization process
EP1286591A1 (en) * 2000-06-07 2003-03-05 Watson Pharmaceuticals, Inc. Treating smooth muscle hyperactivity with (r)-oxybutynin and (r)- desethyloxybutynin
WO2003039464A2 (en) * 2001-11-05 2003-05-15 Pharmacia & Upjohn Company Antimuscarinic aerosol
CN104116760A (en) * 2014-07-14 2014-10-29 玉庆花 An oral liquid for treating enuresis
US10449173B2 (en) 2009-05-05 2019-10-22 Allergan Sales, Llc Method for treating overactive bladders and a device for storage and administration of topical oxybutynin compositions

Families Citing this family (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6171298B1 (en) 1996-05-03 2001-01-09 Situs Corporation Intravesical infuser
US6123961A (en) * 1996-09-25 2000-09-26 Bridge Pharma, Inc. Treating urinary incontinence with (R)-desethyloxybutynin and (R)-oxybutynin
US6130242A (en) * 1997-12-31 2000-10-10 Sepracor Inc. S-procyclidine for treating urinary incontinence
US6183461B1 (en) 1998-03-11 2001-02-06 Situs Corporation Method for delivering a medication
GEP20043354B (en) * 1998-08-27 2004-04-13 Pharmacia & Upjohn Ab Therapeutic Formulation for Administering Tolterodine with Controlled Release
US6432446B2 (en) * 2000-02-03 2002-08-13 Bridge Pharma, Inc. Non-arrhythmogenic metabolite of oxybutynin
US6436428B1 (en) 2000-03-21 2002-08-20 Enhance Pharmaceuticals, Inc. Device and method for treating urinary incontinence in females
PT1280486E (en) 2000-04-26 2011-02-28 Watson Pharmaceuticals Inc Minimizing adverse experience associated with oxybutynin therapy
US7179483B2 (en) 2000-04-26 2007-02-20 Watson Pharmaceuticals, Inc. Compositions and methods for transdermal oxybutynin therapy
US20030124177A1 (en) * 2000-04-26 2003-07-03 Watson Pharmaceuticals, Inc. Compositions and methods for transdermal oxybutynin therapy
US7029694B2 (en) 2000-04-26 2006-04-18 Watson Laboratories, Inc. Compositions and methods for transdermal oxybutynin therapy
AR030312A1 (en) * 2000-08-03 2003-08-20 Antares Pharma Ipl Ag NEW COMPOSITION FOR THE TRANSDERMAL AND / OR TRANSMUCOSICAL ADMINISTRATION OF ACTIVE COMPOUNDS, WHICH ENSURES PROPER THERAPEUTIC LEVELS
US8980290B2 (en) 2000-08-03 2015-03-17 Antares Pharma Ipl Ag Transdermal compositions for anticholinergic agents
US7198801B2 (en) 2000-08-03 2007-04-03 Antares Pharma Ipl Ag Formulations for transdermal or transmucosal application
DE10060550C1 (en) * 2000-12-06 2002-04-18 Lohmann Therapie Syst Lts Transdermal therapeutic system for administration of oxybutynin, especially for treatment of bladder dysfunction, having two-phase matrix layer of active agent-containing droplets dispersed in adhesive
WO2002076426A2 (en) * 2001-03-27 2002-10-03 Galen (Chemicals) Limited Intravaginal drug delivery devices for the administration of an antimicrobial agent
US20030027856A1 (en) * 2001-06-29 2003-02-06 Aberg A.K. Gunnar Tolterodine metabolites
US20030185882A1 (en) * 2001-11-06 2003-10-02 Vergez Juan A. Pharmaceutical compositions containing oxybutynin
US7921999B1 (en) * 2001-12-20 2011-04-12 Watson Laboratories, Inc. Peelable pouch for transdermal patch and method for packaging
AU2002241721A1 (en) * 2001-12-21 2003-09-02 Bridge Pharma, Inc. A non-arrhythmogenic metabolite of oxybutynin
DE60204229T2 (en) * 2002-12-02 2006-02-02 Schwarz Pharma Ag Administration of rotigotine for the treatment of Parkinson's disease by iontophoresis
NZ546106A (en) 2003-10-10 2008-10-31 Antares Pharma Ipl Ag Transdermal pharmaceutical formulation for minimizing skin residues
US7425340B2 (en) * 2004-05-07 2008-09-16 Antares Pharma Ipl Ag Permeation enhancing compositions for anticholinergic agents
WO2007124250A2 (en) 2006-04-21 2007-11-01 Antares Pharma Ipl Ag Methods of treating hot flashes with formulations for transdermal or transmucosal application
WO2006125642A1 (en) 2005-05-27 2006-11-30 Antares Pharma Ipl Ag Methods and apparatus for transdermal or transmucosal application of testosterone
US8415390B2 (en) 2008-05-30 2013-04-09 Microdose Therapeutx, Inc. Methods and compositions for administration of oxybutynin
CA2688542C (en) * 2007-05-30 2016-07-12 Michael J. Martin Methods and compositions for administration of oxybutynin
US9119777B2 (en) 2008-05-30 2015-09-01 Microdose Therapeutx, Inc. Methods and compositions for administration of oxybutynin
NZ607485A (en) 2010-08-03 2015-04-24 Altherx Inc Combinations of beta - 3 adrenergic receptor agonists and muscarinic receptor antagonists for treating overactive bladder
US9907767B2 (en) 2010-08-03 2018-03-06 Velicept Therapeutics, Inc. Pharmaceutical compositions and the treatment of overactive bladder
CN104684549A (en) 2012-02-09 2015-06-03 奥瑟克斯公司 Combination of muscarinic receptor antagonists and beta- 3 adrenoceptor agonists for treating overactive bladder
JP2012176958A (en) * 2012-04-20 2012-09-13 Watson Pharmaceuticals Inc Treatment of smooth muscle hyperactivity by (r)-oxybutynin and (r)-desethyloxybutynin
US20140135392A1 (en) * 2012-11-13 2014-05-15 NeuRx Pharmaceuticals LLC Methods for the treatment of sialorrhea
US20160151321A1 (en) 2012-11-13 2016-06-02 Dinesh C. Patel Methods for the treatment of sialorrhea
KR20170086659A (en) 2014-12-03 2017-07-26 벨리셉트 테라퓨틱스, 인크. Compositions and methods of using modified release solabegron for lower urinary tract symptoms
CN108290824B (en) 2015-10-23 2022-03-11 B3Ar治疗股份有限公司 Sorabegron zwitterion and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE902605A (en) * 1985-06-06 1985-09-30 Therabel Res S A N V Medicaments for treating urinary tract disorders - contg. oxybutynin chloride
JPH06145052A (en) * 1992-11-11 1994-05-24 Hisamitsu Pharmaceut Co Inc Percutaneous administration preparation for treating incontinence of urine
WO1995009007A1 (en) * 1993-09-29 1995-04-06 Alza Corporation Monoglyceride/lactate ester permeation enhancer for oxybutynin

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL267508A (en) * 1960-07-26
PT100502A (en) * 1991-05-20 1993-08-31 Alza Corp PHARMACEUTICAL COMPOSITIONS FOR INCREASING THE CAPACITY OF PERMEACAO IN THE SKIN USING GLYCEROL MONOLINOLEATE

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE902605A (en) * 1985-06-06 1985-09-30 Therabel Res S A N V Medicaments for treating urinary tract disorders - contg. oxybutynin chloride
JPH06145052A (en) * 1992-11-11 1994-05-24 Hisamitsu Pharmaceut Co Inc Percutaneous administration preparation for treating incontinence of urine
WO1995009007A1 (en) * 1993-09-29 1995-04-06 Alza Corporation Monoglyceride/lactate ester permeation enhancer for oxybutynin

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
"Enantiomers of oxybutynin: in vitro pharmacological characterization at M1 m2 and m3 muscarinic receptors and in vivo effects on urinary bladder contraction, mydriasis and salivary secretion in guinea pigs", J.PHARMACOL.EXP.THER., vol. 256, no. 2, 1991, pages 562 - 7, XP000572006 *
"Measurement of oxybutynin and its N-desethyl metabolite in plasma, and its applications to pharmacokinetic studies in young, elderly and frail elderly volunteers", XENOBIOTICA, vol. 22, no. 7, 1992, pages 859 - 69, XP000572170 *
"R and S enantiomers of oxybutynin: pharmacological effects in guinea pig bladder and intestine", J.PHARMACOL.EXP.THER., vol. 247, no. 3, 1988, pages 867 - 72, XP002005849 *
DATABASE WPI Derwent World Patents Index; AN 94-206336, XP002005850 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998000126A1 (en) * 1996-06-28 1998-01-08 Sepracor, Inc. Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin
WO1998011888A1 (en) * 1996-09-19 1998-03-26 American Home Products Corporation Method of treating urinary incontinence
US6013830A (en) * 1998-03-30 2000-01-11 Sepracor Inc. Asymmetric grignard synthesis with cyclic 1,2 aminoalcohols
US6025177A (en) * 1998-03-30 2000-02-15 Sepracor Inc. Asymmetric grignard synthesis with cyclic 1,2 aminoalcohols
WO2000071108A2 (en) * 1999-05-20 2000-11-30 Sepracor Inc. Methods for treatment of asthma using s-oxybutynin
WO2000071108A3 (en) * 1999-05-20 2001-05-17 Sepracor Inc Methods for treatment of asthma using s-oxybutynin
US6294582B1 (en) 1999-05-20 2001-09-25 Sepracor Inc. Methods for treatment of asthma using S-oxybutynin
US6521724B2 (en) 2000-03-10 2003-02-18 E. I. Du Pont De Nemours And Company Polymerization process
EP1286591A4 (en) * 2000-06-07 2007-04-04 Watson Pharmaceuticals Inc Treating smooth muscle hyperactivity with (r)-oxybutynin and (r)- desethyloxybutynin
EP1286591A1 (en) * 2000-06-07 2003-03-05 Watson Pharmaceuticals, Inc. Treating smooth muscle hyperactivity with (r)-oxybutynin and (r)- desethyloxybutynin
DE10103262A1 (en) * 2001-01-25 2002-08-01 Axel Schmidt-Dossi Suppository containing oxybutynin, useful for treatment of incontinence, avoids side effects and allows an increase in dose
WO2002062389A1 (en) * 2001-02-08 2002-08-15 Ono Pharmaceutical Co., Ltd. Remedies for urinary diseases comprising lpa receptor controlling agents
US7288558B2 (en) 2001-02-08 2007-10-30 Ono Pharmaceuticals Co., Ltd. Pharmaceutical composition for treatment for urinary diseases comprising LPA receptor regulator
WO2003039464A3 (en) * 2001-11-05 2004-02-26 Upjohn Co Antimuscarinic aerosol
WO2003039464A2 (en) * 2001-11-05 2003-05-15 Pharmacia & Upjohn Company Antimuscarinic aerosol
US10449173B2 (en) 2009-05-05 2019-10-22 Allergan Sales, Llc Method for treating overactive bladders and a device for storage and administration of topical oxybutynin compositions
CN104116760A (en) * 2014-07-14 2014-10-29 玉庆花 An oral liquid for treating enuresis

Also Published As

Publication number Publication date
CN1179714A (en) 1998-04-22
RU2181589C2 (en) 2002-04-27
UA45387C2 (en) 2002-04-15
MX9705820A (en) 1997-10-31
NZ303372A (en) 1999-01-28
GR3034974T3 (en) 2001-02-28
BR9607001A (en) 1997-10-28
JPH11511730A (en) 1999-10-12
PL182768B1 (en) 2002-02-28
PT806948E (en) 2001-02-28
ATE196252T1 (en) 2000-09-15
CZ242197A3 (en) 1997-12-17
PL321498A1 (en) 1997-12-08
NO315927B1 (en) 2003-11-17
ES2150663T3 (en) 2000-12-01
SK103597A3 (en) 1999-03-12
FI973163A0 (en) 1997-07-30
EP0806948B1 (en) 2000-09-13
US5677346A (en) 1997-10-14
SK283313B6 (en) 2003-05-02
FI973163A (en) 1997-09-30
HUP9800794A2 (en) 1998-07-28
DE69610290T2 (en) 2001-03-29
CA2211400C (en) 2007-01-23
NO973516L (en) 1997-08-07
CZ290093B6 (en) 2002-05-15
CN1151786C (en) 2004-06-02
AU706741B2 (en) 1999-06-24
CA2211400A1 (en) 1996-08-08
KR100390271B1 (en) 2003-10-04
EP0806948A1 (en) 1997-11-19
HK1017986A1 (en) 1999-12-10
DK0806948T3 (en) 2000-12-18
HUP9800794A3 (en) 1999-05-28
NO973516D0 (en) 1997-07-30
DE69610290D1 (en) 2000-10-19
AU4966496A (en) 1996-08-21

Similar Documents

Publication Publication Date Title
EP0806948B1 (en) Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin
AU732568B2 (en) Treating urinary incontinence using (S)-oxybutynin and (S)-desethyloxybutynin
US5532278A (en) Methods and compositions for treating urinary incontinence using optically pure (S)-oxybutynin
US6310103B1 (en) S(−)-tolterodine in the treatment of urinary and gastrointestinal disorders
US6123961A (en) Treating urinary incontinence with (R)-desethyloxybutynin and (R)-oxybutynin
EP1286591B1 (en) Treating smooth muscle hyperactivity with (r)-oxybutynin and (r)- desethyloxybutynin
WO2003066042A1 (en) A non-arrhythmogenic metabolite of oxybutynin
US20030027856A1 (en) Tolterodine metabolites
MXPA97005820A (en) The use of (s) -oxibutinin and (s) -desetiloxibutinin in the preparation of compositions for the treatment of urinary incontinence, the compositions obtained and the procedure for preparing desetiloxibitin
WO1998001125A2 (en) Dextrorotatory isomers of oxybutynin and desethyloxybutynin in the treatment of gastrointestinal hyperactivity
EP1455776A1 (en) A non-arrhythmogenic metabolite of oxybutynin
KR20000022194A (en) Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 96192889.1

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG UZ VN AZ BY KG KZ RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 1996 523636

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2211400

Country of ref document: CA

Ref document number: 2211400

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1996906202

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: PV1997-2421

Country of ref document: CZ

Ref document number: 103597

Country of ref document: SK

WWE Wipo information: entry into national phase

Ref document number: 973163

Country of ref document: FI

Ref document number: 1019970705184

Country of ref document: KR

Ref document number: 303372

Country of ref document: NZ

Ref document number: PA/a/1997/005820

Country of ref document: MX

WWP Wipo information: published in national office

Ref document number: 1996906202

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: PV1997-2421

Country of ref document: CZ

WWP Wipo information: published in national office

Ref document number: 1019970705184

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 1996906202

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: PV1997-2421

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: 1019970705184

Country of ref document: KR