CA2211400C - 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
CA2211400C
CA2211400C CA002211400A CA2211400A CA2211400C CA 2211400 C CA2211400 C CA 2211400C CA 002211400 A CA002211400 A CA 002211400A CA 2211400 A CA2211400 A CA 2211400A CA 2211400 C CA2211400 C CA 2211400C
Authority
CA
Canada
Prior art keywords
oxybutynin
desethyloxybutynin
alpha
less
pharmaceutically acceptable
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.)
Expired - Fee Related
Application number
CA002211400A
Other languages
French (fr)
Other versions
CA2211400A1 (en
Inventor
Gunnar Aberg
John R. Mc Cullough
Yue Fang
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
Priority claimed from US08/381,542 external-priority patent/US5532278A/en
Application filed by Sepracor Inc filed Critical Sepracor Inc
Publication of CA2211400A1 publication Critical patent/CA2211400A1/en
Application granted granted Critical
Publication of CA2211400C publication Critical patent/CA2211400C/en
Anticipated expiration legal-status Critical
Expired - Fee Related 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
    • A61K31/21Esters, 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

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)-DESETHYLOXYBUTYNIN
MELD 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 ]LlIlCtlons or aw.vmoWic: gaiy.mc~.
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. [J.
~harmacol. Ex~- Ther. 256, 562-567 (1991)] concluded that the cholinergic antagonism of racemic oxybutynin (measured in vitro by its affinity for M" MZ 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. ' SUMMARY 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 5' 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 a-cyclohexyl-a-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 a-cyclohexyl-a-hydroxybenzeneacetate, and hereinafter referred to as desethyloxybutynin. The generic name given to the hydrochloride salt of racemic oxybutynin by the USAN

WO 96/23492 PC'yYUS96/01053 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:
,~~~oH
O ° N~
O
I
The S enantiomer of desethyloxybutynin is shown in Formula II:
o ~~~~°H H O
N
O °~
II
The synthesis of (S)-oxybutynin has been described [Kachur et al. J. Pharmacol. EXp. Ther.
~4?, 867-872 (1988)], but (S)-oxybutynin itaelf 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. E_xp- Ther..247, 867-872 (1988) and .
Noronha-Blob et al. J. Pharmacol. ExQ_ Ther. 256, 562-567 (1991)]. (S)-Desethyloxybutynin has not been previously described; its synthesis is carried out according to the method described below.

R'O 96/23492 PCT/US96I01053 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 1 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 a-cyclohexyl-a-hydroxybenzeneacetate with 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butyn-1-of in the presence of an anhydrous base to produce 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butynyl a-cyclohexyl-a-hydroxybenzeneacetate; and then sequentially with a carbonochloridate and methanol to produce 9:-(ethylamino)-2-butynyl a-cyclohexyl-a-hydroxybenzeneacetate (desethyloxybutynin). The process may additionally comprise reacting N-ethyl-4-methoxybenzenemethanamine with 2-propyn-1-of 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-1-of 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.

_7_ Scheme A
OMe Meo ~ OH [CH20]n N-H + HG~
CuCI
HO N
~.-.
V
pH CH3 1 l KzG03 ~ , OH
CDOH acetone GOOMe III IV
NaOMe [cat]
toluene OMe ,OH
O N
C I p O ~, ~ VI
~O~C I
,OH
H 2] MeOH refiux O N
O
VII
The graphic representations of racemic, ambiscalemic and scalemic or enantiomerically pure compounds used herein are taken from Maehr 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) -g-denote enantiomerically pure compounds of indeterminate absolute configuration. a The overall process for DEO involves:
(a) reacting N-ethyl-4-methoxybenzenemethanamine with 2-propyn-1-of and paraformaldehyde in an inert solvent in the presence of cuprous chloride to produce 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butyn-1-of (V);
(b) reacting a single enantiomer of methyl a-cyclohexyl-a-hydroxybenzeneacetate (IV) with 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butyn-1-of (V), in the presence of a catalytic amount of sodium methoxide in toluene to produce a single enantiomer of 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butynyl a-cyclohexyl-a-hydroxybenzeneacetate (VI); and (c) reacting 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butynyl a-cyclohexyl-a-hydroxybenzeneacetate (VI) sequentially with a-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 a-cyclohexyl-a-hydroxybenzeneacetate as well. Paraformal_dehyde 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 a-chloraethyl 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 _g_ 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 responses) 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 WO 96!23492 P~CT/US96/01053 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. Urol.
14$, 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, malefic, 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 5 include suspensions, solutions, elixirs, or solid dosage forms. Carriers such as starches, sugars, and microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like are suitable in the case of oral solid 10 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 1,5 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 20 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
25 92/20377.
Pharmaceutical compositions of the present invention suitable for oral administration may be presented as discrete unit dosage forms such as 30 capsules, cachets, or tablets, each containing a predetermined amount of the active ingredient, as a WO 96!23492 PCT/US96101053 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 pharmaceL~tical art.
Each tablet may contain from about 0.5 mg to about 25 mg of the active ingredient.

R'U 96/23492 PCT/US96/01053 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 tog 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.

WO 96/23492 PCT/~T596/01053 ENANTIOMERS OF OXYBUTYNIN
Binding of !R) - and (S1 -Oxvbutvnin to Human M1,, Mz,,, M3 and M~ Muscarinic Receutor Subty>'es 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 Reference ReceptorRadioligandConc. NonspecificIncubationCompound ' M", (3H)pirenzepine2nM atropine60 min pirenzepine (1pM) 27C

MZH [3HIAF-DX 2nM atropine60 min methoctramine (1NM) 27C

M3H [3H]4-DAMP 0.8nM atropine60 min 4-DAMP

(1pM) 27C

M,H [3H)4-DAMP 0.3nM atropine60min 4-DAMP

(1pM) 27C

Following incubation, the assays were rapidly filtered under vacuum through GF/B glass fiber filters (Whatman) and washed with an ice-cald 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. IC3o 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 SigmaplotTM
software. ICso for R- and S-OXY are given in Table 2.
Table 2 Binding of R-oxybutynin and S-oxybutynin to human muscarinic subtypes M1 - M4 R=OXY S=OXY Ref, Compound Receptor zCja(nM? T~C~(nM).IC3ti(~'1M) M1 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.

R'O 96/23492 fCT/US96101053 Bindina of (R)- and lS)-Oxvbutvnin to Calcium Channels MATERIALS AND METHODS
Bindina assays .
Binding assays were performed using the following methods:
Table 3 Reference Receptors Membranes Compounds References Ca channel rat cerebral diltiazem Schoemaker cortex and (T+L, diltiazem Langer (1985) site) Ca channel rat cerebral D600 Reynolds et cortex al (T+L, verapamil (1986) site) The experiment conditions were:
Table 4 Receptors Ligands ConcentrationsNonsperifiic'Incubatiorr-Ca channel (T+L, diltiazemI'H] diltiazem5nM dilitiazem120 min site) (IO~rM) 25C

Ca channel [3H]D 888 0.5 nM D 600 60 min (T+L,verapamil ' (lOpM) 22C
site) 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).
FXperl.mental Protocols -The compounds were tested in duplicate on each receptor at a concentration of 10-SM. 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.
,~nalvsis 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. ICso 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.
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 ( 10-sM) ( 10-sM) ICso(~) 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 entz-y 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, ICso values were determined for M1 and Mz human receptor subtypes and are presented in Table 6.
Table 6 M, Mist H

10'8M 10''M10'5M ICS 10'9M 10''M 10'5MICS
(nMl (nMl 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 ~~ ~~H
-10'M 10'~M 10'5111110'8M10'7M 10'5M

. ' . S-DEO -- 63 99 -- 43 99 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
,10 SM 10'5M 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 KC1 - to yield a concentration of 137.7 mM KC1 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 KC1 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 , ECso) was estimated for each strip from the second set of concentration-effect data. The ECso 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 ECso of the treated tissue divided by the ECso 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 r 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 AntagonistNo. of pA2 Stope expts:

R-OXY 4 8.80 0.27 1.28 0.26 S-OXY 4 7.09 + 0.13 1.13 + 0.17 RS-OXY 5 8.81 + 0.29 1.34 + 0.15 R-DEO 4 9.04 + 0.32 1.16 + 0.11 S-DEO 4 7.31 0.35 0.87 0.11 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*

'5 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)-a-cyclohexyl-a-hydroxybenzeneacetate (IV) To a mixture of (R)-a-cyclohexyl-a-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 (MeI) (13.0 mL, 208 mmol) dropwise at 0° C (ice bath). After the addition (ca.
1h) of MeI, 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 (8)-a-cyclohexyl-a-hydroxybenzeneacetate (I0):
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).

WO 96!23492 PCT/US96/01053 ~-[N-Ethyl-(~!-methoxyphenyl)methylamino]-2-butynyl (R)-a-cyclohexyl-a-hydroxybenzeneacetate (VI): .
To a solution of (R)-IV(11.9 g, 47.7 mmol) and 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butyn-1-of (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.
~-[N-Ethyl-(4-methoxyphenyl)methylamino]-2-butynyl (8)-a-cyclohexyl-a-hydroxybenzeneacetate (DI):
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 (8)-a-cyclohexyl-a-hydroxybenzene-acetate:
Following the same procedure as above, the racemic precursor to DEO (2.05 g, 43% yield) was obtained as an oil from racsmic IV (2.98 g, 12.0 mmol) and V
(2.48 g, 10.6 mmol).
~1-(Ethylamino)-2-butynyl (R)-a-cyclohexyl-a-hydroxybenzeneacetate hydrochloride salt (VII)-HC1:
A solution of (R)-VI (14.0 g, 31.2 mmol) and a- ' chloroethyl carbonochloridate (4.0 mL, 37.4 mmol) in 1,2-dichloroethane was stirred at reflux for 1 h. r After cooling, the reaction mixture was concentrated and 200 mL of MeOH was added to the residue. The WO 96/23492 PCT/US96l01053 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 . ~ 5 CH2C1~) and then triturated with ether to afford the product (R)-VII-HC1 (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 KZC03 and EtOAc, activated carbon, and a solution of 1N HC1 in ether to give (R)-DEO-HC1 (6.44 g) as an off-white solid.
4-(Ethylamino)-2-butynyl (8)-a-cyclo3iexyl-a-hydroxybenzeneacetate hydrochloride salt (vII)-HCl:
Following the same procedure as above, (S)-DEO-HC1(5.27g, 57% yield) was obtained as an off-white solid from (S)-VI (11.4g, 25.4 mmol).
Racemic 4-(Ethylamino)-2-butynyl a-cyclohexyl-a-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.os mmol).
S-Oxybutynin may be prepared by the same route, substituting 4-(diethylamino)-2-butyn-1-of for the protected intermediate V.
The 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butyn-1-of (V) used as an intermediate is synthesized as follows:
N-Ethyl-~!-methoxybenzenemethanamine:
To a mixture of anisaldehyde (15.6 g, 115 mmol) and ethylamine (2.0 M in THF, 87 mL, 174 mmol) in 1,2-R'O 96/23492 PCT/US96/01053 dichloroethane (450 mL) 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 portianwise 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 CH~Cla 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-1-of A mixture of N-ethyl-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 min. The reaction mixture was cooled to room temperature and concentrated. The residue was 25 diluted with 200 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 CHaClz and then 5% MeOH in CHZC12) to afford the 30 product V (15.1 g, 81% yield) as an oil.

Claims (21)

-29-
1. The use of a compound chosen from the group consisting of (S)-oxybutynin, (S)-desethyloxybutynin and pharmaceutically acceptable salts thereof, wherein the compound contains at least 90% by weight of (S)-oxybutynin or (S)-desethyloxybutynin and 10% by weight or less of (R)-oxybutynin or (R)-desethyloxybutynin, for the manufacture of a medicament for the treatment of urinary incontinence, wherein said use of the compound is from about 1mg to less than 100mg per day, and wherein the treatment is oral, rectal, intravesical, subcutaneous, intramuscular, intravenous, by inhalation, or by aerosol.
2. The use of a compound chosen from the group consisting of (S)-oxybutynin, (S)-desethyloxybutynin and pharmaceutically acceptable salts thereof, wherein the compound contains at least 90% by weight of (S)-oxybutynin or (S)-desethyloxybutynin and 10% by weight or less of (R)-oxybutynin or (R)-desethyloxybutynin, for the treatment of urinary incontinence; wherein said use of the compound is from about 1mg to less than 100mg per day, and wherein the treatment is oral, rectal, intravesical, subcutaneous, intramuscular, intravenous, by inhalation, or by aerosol.
3. The use of (S)-oxybutynin or a pharmaceutically acceptable salt thereof according to claims 1 or 2.
4. The use of (S)-desethyloxybutynin or a pharmaceutically acceptable salt thereof according to claims 1 or 2.
5. The use according to any one of claims 1 to 4 for the use of an oral medicament.
6. A pharmaceutical unit dosage form in the form of a tablet or capsule which comprises from about 0.5mg to less than 100mg of a compound chosen from the group consisting of (S)-oxybutynin and (S)- desethyloxybutynin, or a pharmaceutically acceptable salt thereof, wherein the compound contains at least 90% by weight of (S)-oxybutynin or (S)-desethyloxybutynin and 10% by weight or less of (R)-oxybutynin or (R)-desethyloxybutynin, and a pharmaceutically acceptable carrier.
7. A pharmaceutical unit dosage form according to claim 6 comprising from 0.5 to less than 100 mg of (S)-oxybutynin.
8. A pharmaceutical unit dosage form according to claim 6 comprising from 0.5 to less than 100 mg of (S)-desethyloxybutynin.
9. A process for preparing desethyloxybutynin comprising the steps of:
(a) reacting methyl .alpha.-cyclohexyl-.alpha.hydroxybenzeneacetate with 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butyn-1-ol in the presence of an anhydrous base to produce 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butynyl .alpha.-cyclohexyl-.alpha.-hydroxybenzeneacetate; and (b) reacting said 4-[N-ethyl-(4-methoxyphenyl) methylamino]-2-butynyl .alpha.-cyclohexyl-ahydroxybenzeneacetate sequentially with a carbonochloridate and methanol to produce 4-(ethylamino)-2-butynyl .alpha.-cyclohexyl-.alpha.-hydroxybenzeneacetate.
10. A process according to claim 9 comprising the additional step of reacting N-ethyl-4-methoxybenzenemethanamine with 2-propyn-1-ol and formaldehyde or a formaldehyde equivalent in the presence of a copper(I) salt to produce 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butyn-1-ol.
11. A process according to claim 10 comprising:
(a) reacting N-ethyl-4-methoxybenzenemethanamine with 2-propyn-1-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 .alpha.-cyclohexyl-.alpha.hydroxybenzeneacetate with said 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butyn-1-ol in the presence of sodium methoxide in toluene to produce 4-[N-ethyl-(4-methoxyphenyl)methylamino]-2-butynyl .alpha.-cyclohexyl-.alpha.-hydroxybenzeneacetate; and (c) reacting said 4- [N-ethyl- (4-methoxyphenyl)methylamino]-2-butynyl .alpha.-cyclohexyl-.alpha.-hydroxybenzeneacetate sequentially with .alpha.-chloroethyl carbonochloridate in dichloroethane, followed by methanol to produce 4-(ethylamino)-2-butynyl .alpha.cyclohexyl-.alpha.-hydroxybenzeneacetate(desethyloxybutyin).
12. A process according to claim 9 wherein said desethyloxybutynin is enriched in a single enantiomer.
13. A process according to claim 12 wherein said desethyloxybutynin is S-desethyloxybutynin.
14. The use of a compound chosen from the group consisting of (S)-oxybutynin, (S)-desethyloxybutynin and pharmaceutically acceptable salts thereof, wherein the compound contains at least 99% by weight of (S)-oxybutynin or (S)-desethyloxybutynin and 1% by weight or less of (R)-oxybutynin or (R)-desethyloxybutynin, for the manufacture of a medicament for the treatment of urinary incontinence, wherein said use of the compound is from about 1mg to less than 100mg per day, and wherein the treatment is oral, rectal, intravesical, subcutaneous, intramuscular, intravenous, by inhalation, or by aerosol.
15. The use of a compound chosen from the group consisting of (S)-oxybutynin, (S)-desethyloxybutynin and pharmaceutically acceptable salts thereof, wherein the compound contains at least 99% by weight of (S)-oxybutynin or (S)-desethyloxybutynin and 1% by weight or less of (R)-oxybutynin or (R)-desethyloxybutynin, for the treatment of urinary incontinence, wherein said use of the compound is from about 1mg to less than 100mg per day, and wherein the treatment is oral, rectal, intravesical, subcutaneous, intramuscular, intravenous, by inhalation, or by aerosol.
16. The use of (S) -oxybutynin or a pharmaceutically acceptable salt thereof according to claims 14 or 15.
17. The use of (S)-desethyloxybutynin or a pharmaceutically acceptable salt thereof according to claims 14 or 15.
18. The use according to any one of claims 14 - 17 for the use of an oral medicament.
19. A pharmaceutical unit dosage form in the form of a tablet or capsule which comprises from about 0.5mg to less than 100mg of a compound chosen from the group consisting of (S)-oxybutynin and (S)-desethyloxybutynin, or a pharmaceutically acceptable salt thereof, wherein the compound contains at least 99% by weight of (S)-oxybutynin or (S)-desethyloxybutynin and 1% by weight or less of (R)-oxybutynin or (R)-desethyloxybutynin, and a pharmaceutically acceptable carrier.
20. A pharmaceutical unit dosage form according to claim 19 comprising from 0.5 to less than 100 mg of (S)-oxybutynin.
21. A pharmaceutical unit dosage form according to claim 19 comprising from 0.5 to less than 100 mg of (S)-desethyloxybutynin.
CA002211400A 1995-01-31 1996-01-25 Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin Expired - Fee Related CA2211400C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US08/381,542 US5532278A (en) 1995-01-31 1995-01-31 Methods and compositions for treating urinary incontinence using optically pure (S)-oxybutynin
US08/381,542 1995-01-31
US08/480,194 1995-06-07
US08/480,194 US5677346A (en) 1995-01-31 1995-06-07 Treating urinary incontinence using (S)-desethyloxybutynin
PCT/US1996/001053 WO1996023492A1 (en) 1995-01-31 1996-01-25 Treating urinary incontinence using (s)-oxybutynin and (s)-desethyloxybutynin

Publications (2)

Publication Number Publication Date
CA2211400A1 CA2211400A1 (en) 1996-08-08
CA2211400C true CA2211400C (en) 2007-01-23

Family

ID=27009434

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002211400A Expired - Fee Related CA2211400C (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)

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5736577A (en) * 1995-01-31 1998-04-07 Sepracor, Inc. Methods and compositions for treating urinary incontinence using optically pure (S)-oxybutynin
US6171298B1 (en) 1996-05-03 2001-01-09 Situs Corporation Intravesical infuser
KR20010029519A (en) * 1996-09-19 2001-04-06 이곤 이 버그 Method of treating urinary incontinence
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
US6013830A (en) * 1998-03-30 2000-01-11 Sepracor Inc. Asymmetric grignard synthesis with cyclic 1,2 aminoalcohols
EP1039882B1 (en) * 1998-08-27 2010-11-10 Pfizer Health AB Therapeutic formulation for administering tolterodine with controlled release
CA2371923A1 (en) 1999-05-20 2000-11-30 Thomas P. Jerussi Methods for treatment of asthma using s-oxybutynin
US6432446B2 (en) * 2000-02-03 2002-08-13 Bridge Pharma, Inc. Non-arrhythmogenic metabolite of oxybutynin
US6521724B2 (en) 2000-03-10 2003-02-18 E. I. Du Pont De Nemours And Company Polymerization process
US6436428B1 (en) 2000-03-21 2002-08-20 Enhance Pharmaceuticals, Inc. Device and method for treating urinary incontinence in females
US7029694B2 (en) 2000-04-26 2006-04-18 Watson Laboratories, Inc. Compositions and methods for transdermal oxybutynin therapy
MXPA02010542A (en) * 2000-04-26 2003-10-14 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
ATE483459T1 (en) * 2000-06-07 2010-10-15 Watson Pharmaceuticals Inc TREATING OVERACTIVITY OF SMOOTH MUSCLE WITH (R)-OXYBUTYNIN AND (R)-DESETHYLOXYBUTYNIN
US7198801B2 (en) 2000-08-03 2007-04-03 Antares Pharma Ipl Ag Formulations for transdermal or transmucosal application
ATE485837T1 (en) * 2000-08-03 2010-11-15 Antares Pharma Ipl Ag COMPOSITION FOR TRANSDERMAL AND/OR TRANSMUCOSAL ADMINISTRATION OF ACTIVE INGREDIENTS WHICH GUARANTEES ADEQUATE THERAPEUTIC LEVELS
US8980290B2 (en) 2000-08-03 2015-03-17 Antares Pharma Ipl Ag Transdermal compositions for anticholinergic agents
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
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
JP4396808B2 (en) * 2001-02-08 2010-01-13 小野薬品工業株式会社 Urological disease therapeutic agent comprising LPA receptor modulator
NZ528377A (en) * 2001-03-27 2005-05-27 Galen Chemicals Ltd 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
BR0206300A (en) * 2001-11-05 2008-04-08 Upjohn Co pharmaceutical composition to treat urinary disorder in a mammal and use
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
EP1426049B1 (en) * 2002-12-02 2005-05-18 Schwarz Pharma Ag Iontophoretic delivery of rotigotine for the treatment of Parkinson's disease
MXPA06003316A (en) 2003-10-10 2006-06-08 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
US8067399B2 (en) 2005-05-27 2011-11-29 Antares Pharma Ipl Ag Method 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
US9119777B2 (en) 2008-05-30 2015-09-01 Microdose Therapeutx, Inc. Methods and compositions for administration of oxybutynin
EP2152232A4 (en) * 2007-05-30 2010-06-09 Microdose Therapeutx Inc Methods and compositions for administration of oxybutynin
US8920392B2 (en) 2009-05-05 2014-12-30 Watson Laboratories, Inc. Method for treating overactive bladders and a device for storage and administration of topical oxybutynin compositions
US9907767B2 (en) 2010-08-03 2018-03-06 Velicept Therapeutics, Inc. Pharmaceutical compositions and the treatment of overactive bladder
KR20130135239A (en) 2010-08-03 2013-12-10 앨씨알엑스, 인크. Combinations of beta-3 adrenergic receptor agonists and muscarinic receptor antagonists for treating overactive bladder
SG11201404776PA (en) 2012-02-09 2014-09-26 Altherx Inc 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
US20160151321A1 (en) 2012-11-13 2016-06-02 Dinesh C. Patel Methods for the treatment of sialorrhea
US20140135392A1 (en) * 2012-11-13 2014-05-15 NeuRx Pharmaceuticals LLC Methods for the treatment of sialorrhea
CN104116760A (en) * 2014-07-14 2014-10-29 玉庆花 An oral liquid for treating enuresis
MX2017007054A (en) 2014-12-03 2018-05-02 Velicept Therapeutics Inc Compositions and methods of using modified release solabegron for lower urinary tract symptoms.
FI3365321T3 (en) 2015-10-23 2024-01-02 B3Ar Therapeutics Inc Solabegron zwitterion and uses thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL124473C (en) * 1960-07-26
BE902605A (en) * 1985-06-06 1985-09-30 Therabel Res S A N V Medicaments for treating urinary tract disorders - contg. oxybutynin chloride
PT100502A (en) * 1991-05-20 1993-08-31 Alza Corp PHARMACEUTICAL COMPOSITIONS FOR INCREASING THE CAPACITY OF PERMEACAO IN THE SKIN USING GLYCEROL MONOLINOLEATE
JP3066515B2 (en) * 1992-11-11 2000-07-17 久光製薬株式会社 Transdermal formulation for treatment of urinary incontinence
KR960704579A (en) * 1993-09-29 1996-10-09 에드워드 엘. 만델 Monoglyceride / lactate Ester Permeation Enhancer for Oxybutynin

Also Published As

Publication number Publication date
RU2181589C2 (en) 2002-04-27
JPH11511730A (en) 1999-10-12
AU706741B2 (en) 1999-06-24
SK283313B6 (en) 2003-05-02
DE69610290D1 (en) 2000-10-19
NO315927B1 (en) 2003-11-17
KR100390271B1 (en) 2003-10-04
UA45387C2 (en) 2002-04-15
PL182768B1 (en) 2002-02-28
FI973163A (en) 1997-09-30
ES2150663T3 (en) 2000-12-01
BR9607001A (en) 1997-10-28
NO973516D0 (en) 1997-07-30
NZ303372A (en) 1999-01-28
HUP9800794A2 (en) 1998-07-28
MX9705820A (en) 1997-10-31
CZ290093B6 (en) 2002-05-15
PT806948E (en) 2001-02-28
HK1017986A1 (en) 1999-12-10
EP0806948B1 (en) 2000-09-13
ATE196252T1 (en) 2000-09-15
SK103597A3 (en) 1999-03-12
CN1179714A (en) 1998-04-22
DK0806948T3 (en) 2000-12-18
DE69610290T2 (en) 2001-03-29
GR3034974T3 (en) 2001-02-28
CZ242197A3 (en) 1997-12-17
NO973516L (en) 1997-08-07
US5677346A (en) 1997-10-14
AU4966496A (en) 1996-08-21
EP0806948A1 (en) 1997-11-19
HUP9800794A3 (en) 1999-05-28
FI973163A0 (en) 1997-07-30
WO1996023492A1 (en) 1996-08-08
PL321498A1 (en) 1997-12-08
CN1151786C (en) 2004-06-02
CA2211400A1 (en) 1996-08-08

Similar Documents

Publication Publication Date Title
CA2211400C (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
AU728395B2 (en) S(-)-tolterodine in the treatment of urinary and gastrointestinal disorders
US6123961A (en) Treating urinary incontinence with (R)-desethyloxybutynin and (R)-oxybutynin
AU2000255966B2 (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
KR100476550B1 (en) Urinary incontinence treatment with (S) -oxybutynin and (S) -desethyloxybutynin
WO1998001125A2 (en) Dextrorotatory isomers of oxybutynin and desethyloxybutynin in the treatment of gastrointestinal hyperactivity
EP1455776A1 (en) A non-arrhythmogenic metabolite of oxybutynin
JP2005516992A6 (en) Non-cardiac rhythm-inducing metabolite of oxybutynin

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed