WO1992009560A1 - Gaba and l-glutamic acid analogs for antiseizure treatment - Google Patents

Gaba and l-glutamic acid analogs for antiseizure treatment Download PDF

Info

Publication number
WO1992009560A1
WO1992009560A1 PCT/US1991/008701 US9108701W WO9209560A1 WO 1992009560 A1 WO1992009560 A1 WO 1992009560A1 US 9108701 W US9108701 W US 9108701W WO 9209560 A1 WO9209560 A1 WO 9209560A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
set forth
hydrogen
administering
gaba
Prior art date
Application number
PCT/US1991/008701
Other languages
French (fr)
Inventor
Richard B. Silverman
Original Assignee
Northwestern University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Northwestern University filed Critical Northwestern University
Publication of WO1992009560A1 publication Critical patent/WO1992009560A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/04Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C229/06Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton
    • C07C229/08Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one amino and one carboxyl group bound to the carbon skeleton the nitrogen atom of the amino group being further bound to hydrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/04Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C229/24Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having more than one carboxyl group bound to the carbon skeleton, e.g. aspartic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/28Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being saturated and containing rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/34Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/66Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety
    • C07C69/67Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety of saturated acids
    • C07C69/675Esters of carboxylic acids having esterified carboxylic groups bound to acyclic carbon atoms and having any of the groups OH, O—metal, —CHO, keto, ether, acyloxy, groups, groups, or in the acid moiety of saturated acids of saturated hydroxy-carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated

Definitions

  • the present invention relates to novel compounds that are analogs of glutamic acid and gamma-aminobutyric acid (GABA). More specifically, the analogs are useful as antiseizure therapy for central nervous system disorders such as epilepsy, Huntington's chorea, cerebral ischemia, Parkinson's disease, tardive dyskinesia and spasticity. It is also possible that the present invention could be used as an anti-depressant, anxiolytic and
  • GABA Gamma-aminobutyric acid
  • L-glutamic acid are two major neurotransmitters
  • GABA is the major inhibitory
  • neurotransmitter and L-glutamic acid is an excitatory transmitter (1,2).
  • An imbalance in the concentration of these neurotransmitters can lead to convulsive states. Accordingly, it is clinically relevant to be able to control convulsive states by controlling the metabolism of this neurotransmitter.
  • the tern seizure as used herein means excessive unsynchronized neuronal activity that disrupts normal neuronal function. In several seizure disorders there is concomitant with reduced brain GABA levels a
  • GAD L-glutamic acid decarboxylase
  • GABA As an inhibitory neurotransmitter, and its effect on convulsive states and other motor dysfunctions, a variety of approaches have been taken to increase the brain GABA concentration. For example, the most obvious approach was to administer GABA. When GABA is injected into the brain of a convulsing animal, the convulsions cease (10). However, if GABA is administered systemically, there is no anticonvulsant effect because GABA, under normal circumstances, cannot cross the blood brain barrier (11). In view of this limitation, there are three alternative approaches that can be taken to raise GABA levels.
  • GABA lipophilic by making GABA lipophilic by conversion to hydrophobic GABA amides (13,14), imines (13), or GABA esters (15,16) so that GABA can cross the blood brain barrier.
  • GABA esters Once inside the brain, these compounds require amidases and esterases to hydrolyze off the carrier group and release GABA.
  • a third approach is to increase brain GABA levels by designing an activator of GAD.
  • GAD GABA-dependent neuropeptide
  • the anticonvulsant agent, milacemide was reported to increase the activity of GAD by 11% and as a result increase GABA concentration in the substantia nigra by up to 38% (17).
  • the anticonvulsant drug sodium valproate (18) was also reported to activate GAD and increase GABA levels.
  • Applicant has synthesized a series of GABA and L-glutamate analogs having the ability to activate GAD in vitro and having a dose dependent protective effect of seizure in vivo.
  • One compound in particular was found to be an unexpectedly potent suppressor of seizures while the entire series of drugs do not promote the unwanted side effects of ataxia. Accordingly, the present invention provides a novel series of compounds and their method of use in suppressing seizures.
  • R 1 is a straight or branched alkyl of from 1 to 6 carbons, phenyl or cycloalkyl having from 3 to 6 carbon atoms
  • R 2 is hydrogen or methyl
  • R 3 is hydrogen, methyl or carboxyl; or its diastereomers, or enantiomers and pharmaceutically acceptable salts thereof.
  • the present invention further provides a method of treating seizure disorders by administering an anticonvulsant effective amount of the
  • the present invention provides a method for increasing brain neuronal GABA and
  • the alkyl moieties as represented by R 1 in Formula I can be methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, isopentyl and neopentyl as well as other alkyl groups.
  • the cycloalkyl groups represented by R 1 are exemplified by cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • the analogs are further shown herein to prevent seizure while not causing the side effect of ataxia, such a side effect being found in several anti-seizure pharmaceuticals. More specifically, the present invention provides compounds of the formula I
  • R 1 is a straight or branched alkyl of from 1 to 6 carbons, phenyl or a cyloalkyl having 3 to 6 carbon atoms
  • R 2 is hydrogen or methyl
  • R 3 is hydrogen, methyl, or carboxyl; or its diastereomers; or enantiomers, and both pharmaceutically acceptable salts thereof.
  • the most preferred compounds of the present invention are of the formula above wherein R 3 is hydrogen, R 2 is hydrogen and R 1 isobutyl. That is, the preferred compound is 4-amino-3-(2-methylpropyl) butanoic acid. It has been found that this compound is unexpectedly more potent than the other analogs synthesized in accordance herewith and tested in vivo.
  • this preferred compound is the least effective one of the analogs tested in activating GAD in vitro. Accordingly, it was very unexpected that this preferred compound had such a high potentency when tested in vivo.
  • the compounds made in accordance with the present invention may form pharmaceutically
  • the acid addition salts of the basic compounds are prepared either by
  • salts examples include hydrochlorides, hydrobromides, hydrosulfates, etc. as well as sodium, potassium and magnesium etc. salts.
  • the compounds made in accordance with the present invention can contain one or several
  • the invention includes the individual diastereomers or enantiomers, and the mixtures thereof.
  • the individual diastereomers or enantiomers may be prepared or isolated by methods already well known in the art.
  • the method for the formation of the 3-alkyl-4-aminobutanoic acids starting from 2-alkenoic esters is prepared from commercially available aldehydes and monoethyl malonate by the Knoevenagel reaction (19), with the exception of ethyl 4,4-dimethyl-2-pentenoate.
  • This compound was prepared from 2,2-dimethylpropanal and ethyl lithioacetate, followed by dehydration of the beta-hydroxyester with phosphoryl chloride and pyridine.
  • the compounds made by the aforementioned synthetic method can be used as pharmaceutical compositions as an anti-depressant, anxiolytic, antipsychotic, antiseizure, anti-dyskinesic, or anti-symptomatic for Huntington's or Parkinson's diseases when an effective amount of a compound of the aforementioned formula together with a
  • the present invention provides a pharmaceutical composition for the suppression of seizures resulting from epilepsy, the treatment of cerebral ischemia, Parkinson's disease, Huntington's disease and
  • the pharmaceutical can be used in a method for treating such disorders in mammals, including human, suffering therefrom by administering to such mammals an effective amount of the compound as described above in unit dosage form.
  • compositions can be made in inert, pharmaceutically acceptable carriers which are either solid or liquid.
  • solid form
  • preparation include powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • the quantity of active compound in a unit dose of preparation may be varied or adjusted from one milligram to about 300 milligrams per kilogram daily, based on an average 70 kilogram patient. A daily dose range of about 1 milligram to about 50 milligrams per kilogram is preferred. The dosages, however, may be varied depending upon the requirement with a patient, the severity of the condition being treated, and the compound being employed.
  • L-glutamic acid 0.5, 0.25, 0.166, 0.125, 0.10 mM
  • [ 14 C]L-glutamate (10 ⁇ C i /mmol) in 50 mM potassium phosphate buffer, pH 7.2 were shaken at 37°C in separate vials with purified L-glutamic acid decarboxylase (18.75 ⁇ g; spec, act 10.85 jumol/min rag) in a total volume of 2.00 ml.
  • the enzyme reactions were quenched by the addition of 200 ⁇ l of 6 M sulfuric acid to the contents of each of the vials.
  • the vials were shaken for an additional 60 minutes at 37°C.
  • the center wells were removed and placed in scintillation vials with 10 ml of scintillation fluid for radioactivity determination.
  • the same assays were repeated except in the presence of various concentrations of the activators (2.5, 1.0, 0.5, 0.25, 0.1, 0.05 mM).
  • Vmax values were determined from plots of 1/cpm versus 1/[glutamate] at various concentrations of activators. The data were expressed as the ratio of the V max in the presence of the activators to the
  • Threshold maximum electroshock is an animal model test for generalized seizures that is similar to that of Piredda, S.G. et al (21). The methods for this test are described as follows.
  • mice Male CF-1 mice (22-30 grams) were allowed free access to food and water prior to testing. For screening, groups of five mice were given a compound intravenously at doses of 30, 100, and 300 mg/kg and tested at 0.5, 2.0 and 4.0 hours after dosing. Drugs were either dissolved in 0.9% saline or suspended in 0.2% methylcellulose. Animals were shocked with corneal electrodes (see below) and observed for tonic hindlimb extensor seizures. Absence of hindlimb extension was taken as an anticovulsant effect.
  • the electroshock apparatus delivered a 60 Hz sine wave with a current amplitude of 14 mA
  • electroshock tests where conducted varying the time of testing from one hour to eight hours, the dose being 10 milligram per kilogram in mice, injected intravenously. Table 3 shows the results of these tests indicating a maximum protection after two hours of testing.
  • inventions are of value as pharmacological agents, particularly for the treatement of seizures in mammals, including humans.
  • High-intensity corneal electroshock consisted of 50 mA, base-to-peak sinusoidal current for 0.2 sec. All other data was from low-intensity electroshock, 17 mA base-to-peak sinusoidal current for 0.2 sec.

Abstract

Novel analogs of GABA and L-glutamic acid are used for treating seizure disorders. One analog, 4-amino-3-(2-methylpropyl) butanoic acid is found to have unexpectedly potent antiseizure activity in vivo.

Description

GABA AND L-GLUTAMIC ACID ANALOGS
FOR ANTISEIZURE TREATMENT
TECHNICAL FIELD
The present invention relates to novel compounds that are analogs of glutamic acid and gamma-aminobutyric acid (GABA). More specifically, the analogs are useful as antiseizure therapy for central nervous system disorders such as epilepsy, Huntington's chorea, cerebral ischemia, Parkinson's disease, tardive dyskinesia and spasticity. It is also possible that the present invention could be used as an anti-depressant, anxiolytic and
antipsychotic activity.
BACKGROUND OF THE INVENTION
Gamma-aminobutyric acid (GABA) and L-glutamic acid are two major neurotransmitters
involved in the regulation of brain neuronal
activity. GABA is the major inhibitory
neurotransmitter and L-glutamic acid is an excitatory transmitter (1,2). An imbalance in the concentration of these neurotransmitters can lead to convulsive states. Accordingly, it is clinically relevant to be able to control convulsive states by controlling the metabolism of this neurotransmitter.
When the concentration of GABA diminishes below a threshold level in the brain, convulsions result (3); when the GABA levels rise in the brain during convulsions, seizures terminate (4). The tern seizure as used herein means excessive unsynchronized neuronal activity that disrupts normal neuronal function. In several seizure disorders there is concomitant with reduced brain GABA levels a
diminished level of L-glutamic acid decarboxylase (GAD) activity also observed (5-9). Often, the concentrations of GAD and GABA vary in parallel because decreased GAD concentration results in lower GABA production.
Because of the importance of GABA as an inhibitory neurotransmitter, and its effect on convulsive states and other motor dysfunctions, a variety of approaches have been taken to increase the brain GABA concentration. For example, the most obvious approach was to administer GABA. When GABA is injected into the brain of a convulsing animal, the convulsions cease (10). However, if GABA is administered systemically, there is no anticonvulsant effect because GABA, under normal circumstances, cannot cross the blood brain barrier (11). In view of this limitation, there are three alternative approaches that can be taken to raise GABA levels.
The most frequent approach is to design a compound that crosses the blood brain barrier and then inactivates GABA aminotransferase. The effect is to block the degradation of GABA and thereby increase its concentration. Numerous mechanism-based inactivators of GABA aminotransferase are known (12).
Another approach is to increase GABA concentrations in the brain by making GABA lipophilic by conversion to hydrophobic GABA amides (13,14), imines (13), or GABA esters (15,16) so that GABA can cross the blood brain barrier. Once inside the brain, these compounds require amidases and esterases to hydrolyze off the carrier group and release GABA.
A third approach is to increase brain GABA levels by designing an activator of GAD. A few compounds have been described as activators of GAD. The anticonvulsant agent, milacemide, was reported to increase the activity of GAD by 11% and as a result increase GABA concentration in the substantia nigra by up to 38% (17). The anticonvulsant drug sodium valproate (18) was also reported to activate GAD and increase GABA levels. Applicant has synthesized a series of GABA and L-glutamate analogs having the ability to activate GAD in vitro and having a dose dependent protective effect of seizure in vivo. One compound in particular was found to be an unexpectedly potent suppressor of seizures while the entire series of drugs do not promote the unwanted side effects of ataxia. Accordingly, the present invention provides a novel series of compounds and their method of use in suppressing seizures.
SUMMARY OF THE INVENTION
In accordance with the present invention, there is provided a compound of the formula I
Figure imgf000006_0001
wherein R1 is a straight or branched alkyl of from 1 to 6 carbons, phenyl or cycloalkyl having from 3 to 6 carbon atoms, R2 is hydrogen or methyl, and R3 is hydrogen, methyl or carboxyl; or its diastereomers, or enantiomers and pharmaceutically acceptable salts thereof. The present invention further provides a method of treating seizure disorders by administering an anticonvulsant effective amount of the
aformentioned composition.
Also, the present invention provides a method for increasing brain neuronal GABA and
provides pharmaceutical compositions of the compounds of Formula I. DETAILED DESCRIPTION OF THE INVENTION
In accordance with the present invention, there is provided a series of 3-alkyl-4-aminobutyric acid or 3-alkyl glutamic acid analogs which are shown herein to activate GAD. For example, the alkyl moieties as represented by R1 in Formula I can be methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, isopentyl and neopentyl as well as other alkyl groups. The cycloalkyl groups represented by R1 are exemplified by cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The analogs are further shown herein to prevent seizure while not causing the side effect of ataxia, such a side effect being found in several anti-seizure pharmaceuticals. More specifically, the present invention provides compounds of the formula I
Figure imgf000008_0001
wherein R1 is a straight or branched alkyl of from 1 to 6 carbons, phenyl or a cyloalkyl having 3 to 6 carbon atoms R2 is hydrogen or methyl, and R3 is hydrogen, methyl, or carboxyl; or its diastereomers; or enantiomers, and both pharmaceutically acceptable salts thereof. The most preferred compounds of the present invention are of the formula above wherein R3 is hydrogen, R2 is hydrogen and R1 isobutyl. That is, the preferred compound is 4-amino-3-(2-methylpropyl) butanoic acid. It has been found that this compound is unexpectedly more potent than the other analogs synthesized in accordance herewith and tested in vivo. What is further surprising, as the following data shows, is that this preferred compound is the least effective one of the analogs tested in activating GAD in vitro. Accordingly, it was very unexpected that this preferred compound had such a high potentency when tested in vivo. The compounds made in accordance with the present invention may form pharmaceutically
acceptable salts with both organic and inorganic acids or bases. For example, the acid addition salts of the basic compounds are prepared either by
dissolving the free base in aqueous or aqueous alcohol solution or other suitable solvents
containing the appropriate acid and isolating the salt by evaporating the solution. Examples of pharmaceutically acceptable salts are hydrochlorides, hydrobromides, hydrosulfates, etc. as well as sodium, potassium and magnesium etc. salts.
The compounds made in accordance with the present invention can contain one or several
asymmetric carbon atoms. The invention includes the individual diastereomers or enantiomers, and the mixtures thereof. The individual diastereomers or enantiomers may be prepared or isolated by methods already well known in the art.
The method for the formation of the 3-alkyl-4-aminobutanoic acids starting from 2-alkenoic esters is prepared from commercially available aldehydes and monoethyl malonate by the Knoevenagel reaction (19), with the exception of ethyl 4,4-dimethyl-2-pentenoate. This compound was prepared from 2,2-dimethylpropanal and ethyl lithioacetate, followed by dehydration of the beta-hydroxyester with phosphoryl chloride and pyridine. The Michael addition of nitromethane to alpha, beta-unsaturated compounds mediated by 1,1,3,3-tetramethylguanidine or 1,8-diazabicyclo[5.4.0]undec-7-ene(DBU) afforded 4- nitroesters in good yields.
Although the aliphatic nitro compounds are usually reduced by either high pressure catalyic hydrogenation by metal-catalyzed transfer
hydrogenation, or by newly introduced hydrogenolysis methods with ammonium formate or sodium borohydride and palladium as catalysts , applicants have found that 4-nitrocarboxylic esters can be reduced almost quantatitively to the corresponding 4-aminocarboxylic esters by hydrogenation using 10% palladium on carbon as catalysts in acetic acid at room temperature and atmospheric pressure. The amino esters produced were subjected to acid hydrolysis to afford the subject inventive compounds in good yields. This procedure provides access to a variety of 3-alkyl-4-aminobutanoic acids as listed in Tables 1 and 2 as examples and thus is advantageous in comparison to methods previously used. When the starting material is not
commercially available, the synthetic sequence was initiated with the corresponding alcohol, which was oxidized to the aldehyde by the method of Corey et al (20).
The compounds made by the aforementioned synthetic method can be used as pharmaceutical compositions as an anti-depressant, anxiolytic, antipsychotic, antiseizure, anti-dyskinesic, or anti-symptomatic for Huntington's or Parkinson's diseases when an effective amount of a compound of the aforementioned formula together with a
pharmaceutically acceptable carrier is used. That is, the present invention provides a pharmaceutical composition for the suppression of seizures resulting from epilepsy, the treatment of cerebral ischemia, Parkinson's disease, Huntington's disease and
spasticity and also possibly for antidepressent, anxiolytic and antipsychotic effects. These latter uses are. expected due to functional similarities to other known compounds having these pharmacological activities. The pharmaceutical can be used in a method for treating such disorders in mammals, including human, suffering therefrom by administering to such mammals an effective amount of the compound as described above in unit dosage form. The pharmaceutical compound made in
accordance with the present invention can be prepared and administered in a wide variety of dosage forms. For example, these pharmaceutical compositions can be made in inert, pharmaceutically acceptable carriers which are either solid or liquid. Solid form
preparation include powders, tablets, dispersible granules, capsules, cachets, and suppositories.
Other solid and liquid form preparations could be made in accordance with known methods of the art.
The quantity of active compound in a unit dose of preparation may be varied or adjusted from one milligram to about 300 milligrams per kilogram daily, based on an average 70 kilogram patient. A daily dose range of about 1 milligram to about 50 milligrams per kilogram is preferred. The dosages, however, may be varied depending upon the requirement with a patient, the severity of the condition being treated, and the compound being employed.
Determination of the proper dosage for particular situations is within the skill of the art.
Illustrative examples of compounds made in accordance with the present invention were tested to demonstrate the ability of the compounds to activate GAD in vitro and to prevent seizure in vivo without the side effect of ataxia. In Vitro GAD Activation
Assays were carried out in 10 ml vials sealed with serum caps through which a center well (Kontes catalogue no. 882320-000) was inserted. The center well was charged with 200 μl of freshly prepared 8% KOH solution. Various concentrations of
L-glutamic acid (0.5, 0.25, 0.166, 0.125, 0.10 mM) containing [14C]L-glutamate (10 μCi/mmol) in 50 mM potassium phosphate buffer, pH 7.2 were shaken at 37°C in separate vials with purified L-glutamic acid decarboxylase (18.75 μg; spec, act 10.85 jumol/min rag) in a total volume of 2.00 ml. After being shaken for
60 minutes, the enzyme reactions were quenched by the addition of 200 μl of 6 M sulfuric acid to the contents of each of the vials. The vials were shaken for an additional 60 minutes at 37°C. The center wells were removed and placed in scintillation vials with 10 ml of scintillation fluid for radioactivity determination. The same assays were repeated except in the presence of various concentrations of the activators (2.5, 1.0, 0.5, 0.25, 0.1, 0.05 mM). The
Vmax values were determined from plots of 1/cpm versus 1/[glutamate] at various concentrations of activators. The data were expressed as the ratio of the Vmax in the presence of the activators to the
Vmax in the absence of the activators times 100%. The results of the experiment are shown in Table 1. The tests show that there was significant activation by the various compounds tested to differing degrees. The known activator sodium valproate and GABAPENTIN were tested.
In vivo tests were performed to demonstrate the seizure preventing capabilities of the novel compounds. Threshold maximum electroshock is an animal model test for generalized seizures that is similar to that of Piredda, S.G. et al (21). The methods for this test are described as follows.
Male CF-1 mice (22-30 grams) were allowed free access to food and water prior to testing. For screening, groups of five mice were given a compound intravenously at doses of 30, 100, and 300 mg/kg and tested at 0.5, 2.0 and 4.0 hours after dosing. Drugs were either dissolved in 0.9% saline or suspended in 0.2% methylcellulose. Animals were shocked with corneal electrodes (see below) and observed for tonic hindlimb extensor seizures. Absence of hindlimb extension was taken as an anticovulsant effect.
The electroshock apparatus delivered a 60 Hz sine wave with a current amplitude of 14 mA
(peak-to-peak) for 0.2 seconds. The current strength of 14 mA used in this procedure produced tonic extensor seizures in approximately 95% of untreated mice, but was only slightly above threshold for tonic extension.
Summaries of the numbers of animals
protected from seizures when tested 120 minutes after administration of each compound set forth in the lefthand column are given in Table 2 for varying dose levels set forth in the second column of the Table.
Due to the interesting phenomena related to the (R,S)-i-butyl GABA (the compound having
significantly higher potency and effectiveness without causing ataxia), threshold maximal
electroshock tests where conducted varying the time of testing from one hour to eight hours, the dose being 10 milligram per kilogram in mice, injected intravenously. Table 3 shows the results of these tests indicating a maximum protection after two hours of testing.
In view of the above results, a dose response curve was made for the two hour testing time period in mice, the drug being given intravenously at 10 milligrams per kilogram. The results of this test is shown in Table 4 with a calculated ED50 equaling 2.07 milligrams per kilogram. A third pharmacological test was performed as described in R.L. Krall et al (22). In this procedure, drugs were tested for attenuation of threshold clonic seizures in mice caused by
subcutaneous administration of pentylenetetrazol (85 mg/kg) which is a generally accepted model for absence type seizures. Results from the third test for the compound when administered either
intravenously or orally is shown in Table 5. The test was conducted at three dose levels, showing effective protection at 30 mg/kg and 100 mg/kg with no ataxia.
The above is a significant finding because the compound having the least ability to activate GAD surprisingly had an approximately 10 fold increase in potency over the other compounds tested. Even more unexpected is the absence of ataxic side effect coupled to this increase in potency.
In view of the above demonstrated activity of the compounds characterizing the present invention and in particular the 4-amino-3-(2-methylpropyl)butanoic acid (isobutyl GABA) the compounds made in accordance with the present
invention are of value as pharmacological agents, particularly for the treatement of seizures in mammals, including humans.
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000018_0002
TABLE 2
Prevention of tonic extensor seizures in mice following intravenous administration of 3-substituted GABA derivatives.
dose time after effect ataxia
R (mg/kg) dose (min) #protected/ 3 ataxia
# tested # tested
(R,S)-CH3 10 120 0/5 0/5
30 120 4/5 0/5
100 120 3/5 0/5
(R)-CH3 1 120 1/10 0/10
3 120 2/10 0/10
10 120 4/10 0/10
30 120 3/10 0/10
100 120 3/10(5/10) 1/10
(S)-CH3 10 120 1/10 1/10
30 120 2/10 0/10
100 120 5/10 0/10 t-C4H9 10 120 2/10 0/10
30 120 2/10 0/10
100 120 5/10 0/10
C2H5 3 120 1/5 0/5
10 120 1/5 0/5
30 120 2/5 0/5
100 120 5/5 0/5
(CH3)2 30 120 4/5 0/5
100 120 4/5 0/5 n-C4H9 10 120 1/10 0/10
30 120 3/10 0/10
100 120 4/10 0/10
3 120 2/10 0/10 s-C4H9 10 120 3/10 0/10
30 120 2/10 0/10 i-C4H9 0.3 120 1/10 0/10
0.8 120 3/10 0/10 2.0 120 5/10 0/10
5.5 120 7/10 0/10 14.4 120 9/10 0/10 n-C3H7 3 120 2/10 0/10
10 120 2/10 3/10
100 120 3/10 0/10 i-C3H7 10 120 5/10 1/10
30 120 5/10 0/10
100 120 6/10 0/ 10 C6H5 5 100 120 0/10 0/10 neo-C5H11 10 120 2/10 0/10
30 120 4/10 0/10
100 120 4/10 0/10
- High-intensity corneal electroshock consisted of 50 mA, base-to-peak sinusoidal current for 0.2 sec. All other data was from low-intensity electroshock, 17 mA base-to-peak sinusoidal current for 0.2 sec.
TABLE 3
Threshold Maximal Electroshock with Isobutyl GABA.
Time of Testing # Protected
1 hr. 2/10
2 hr. 8/10
4 hr. 4/10
8 hr. 2/10
TABLE 4
Dose m/k # Protected
0.3 1/10
0.8 3/10
2.0 5/10
5.5 7/10
14.4 9/10
TABLE 5
Maximal electroshock data:
dose time after effect ataxia
R (mg/kg) dose (min) # protected/ # ataxic
# tested # tested i-C4H9 10 120 1/5 0/5 i-C4H9 30 120 4/5 0/5 i-C4H9 100 120 4/5 0/5
REFERENCES
1. Roberts, E. et al, GABA in Nervous System Fucntion;
Raven Press: New York, 1976.
2. McGeer, E.G. et al, Glutamine, Glutamate, and GABA in the Central Nervous System; Hertz, L. ; Kvamme, E.;
McGeer, E.G.; Schousbal, A., Eds.: Liss: New York, 1983, pp. 3-17.
3. Karlsson, A. et al, Biochem. Pharmacol 1974, 23, 3053- 3061.
4. Hayashi, T.J., Physiol. (London) 1959, 145, 570-578.
5. McGeer, P.O. et al. In GAGA in Nervous System Function;
Roberts, E.; Chase, T.N.; Tower, D.B., Eds.; Raven: New York 1976; pp. 487-495.
6. Butterworth, J. et al, Neurochem. 1983, 41, 440-447.
7. Spokes, E.G., Adv. Exp. Med. Biol. 1978, 123, 461-473.
8. Wu, J.Y. et al, Neurochem. Res. 1979, 4, 575-586.
9. Iversen, L.L. et al, Psychiat. Res. 1974, 11, 255-256.
10. Purpura, D.P. et al, Nurochem, 1959, 3, 238-268.
11. Meldrum, B.S. et al. Epilepsy; Harris, P., Mawdsley,
C.., Eds.; Churchill Livingston: Edinburg; 1974, p. 55.
12. Silverman, R.B., Mechanism-Based Enzyme Inactivation:
Chemistry and Enzymology, Vol. I and II; CRC: Boca
Raton; 1988.
13. Kaplan, J.P. et al; G.J. Med. Chem. 1980, 23, 702-704.
14. Carvajal, G. et al; Biochem. Pharmacol. 1964, 13, 1059- 1069.
15. Shashoua, V.E. et al; J. Med. Chem. 1984, 27, 659-664.
16. PCT Patent Application WO85/00520, published 2/14/85.
17. Janssens de Varebeke, P. et al, Biochem. Pharmacol 1933, 32, 2751-2755.
18. Loscher, W., Biochem. Pharmacol 1982, 31, 837-842;
Phillips, N.I. et al, Biochem. Pharmacol. 1982, 31, 2257-2261. References
19. Kim, Y.C., Cocolase G.H., J. Med. Chem. 1965,
8509.
20. Corey, E.J. et al, Tetrahedrom Lett. 1975, 2647-2650.
21. Piredda, S.G. et al, Effect of Stimulus Intensity on the Profile of Anticonvulsant Activity of Phenytoin,
Ethosuximide and Valproate, The Journal of Pharmacology and Experimental Therapeutics, 232, (3):741-45 (1985).
22. Krall, R.L. et al, Epilepsia. 19:409 (1978).

Claims

What is claimed is:
1. A compound of the formula
Figure imgf000024_0001
wherein R1 is a straight or branched alkyl of from one to six carbons, a phenyl, or a cycloalkyl having from 3 to 6 carbon atoms , R2 is hydrogen or methyl, and R3 is hydrogen; methyl or carboxyl;. or its diastereomers; or enantiomers; and both
pharmaceutically acceptable salts thereof.
2. A compound as set forth in claim 1 wherein R3 is hydrogen, R2 is hydrogen, and R1 is
-(CH2)n-i C4H9 as an (R), (S) or (R,S) isomer, n being 0-2.
3. A compound as set forth in claim 2 and being 4-amino-3-(2-methylpropyl)butanoic acid.
4. The use of a compound for treating seizure disorders in a patient which comprises administering to the patient an effective amount of the compound of the formula
Figure imgf000025_0002
wherein R is an isobutyl; n is zero, one, or two and its enantiomers or a pharmaceutically acceptable salt thereof.
5. The use of a compound as set forth in claim 4 wherein the compound is 4-amino-3-(2-methylpropyl)butanoic acid or a pharmaceutically acceptable salt thereof.
6. A method of treating seizure disorders in a patient, said method including the steps of administering an anticonvulsant effective amount of a composition having the formula
Figure imgf000025_0001
wherein R1 is a straight or branched alkyl of from one to six carbons, a phenyl or a cycloalkyl of 3 to 6 carbons , R2 is hydrogen or methyl, and R3 is hydrogen, methyl or carboxyl; or its diastereomers; or enantiomers; and pharmaceutically acceptable salt. thereof.
7. A method as set forth in claim 6 wherein the compound is 4-amino-3-(2- methylpropyl)butanoic acid.
8. A method as set forth in claim 6 wherein said administering step is further defined as orally administering the compound.
9. A method as set forth in claim 4 wherein said administering step is further defined as intravenously administering the compound.
10. A pharmaceutical composition for treating seizures comprising an anticonvulsant effective amount of a compound of the formula
Figure imgf000026_0001
wherein R1 is a straight chain or branched alkyl of from one to six carbons, a phenyl or a cycloalkyl of three to six carbons, R2 is hydrogen or methyl, and R3 is hydrogen, methyl or carboxyl; or its
diastereomers; or enantiomers; and pharmaceutically acceptable salts thereof and pharmaecutical carrier.
11. A pharmaceutical as set forth in claim
10 wherein R3 is hydrogen, R2 is hydrogen, and R1 is -(CH2)n-i C4H9 as an (R), (S) or (R,S) isomer, n being 0-2.
12. A pharmaceutical as set forth in claim
11 wherein the active ingredient is 4-amino-3-(2-methylpropyl) butanoic acid.
13. A method of increasing brain neuronal
GABA levels, said method including the steps of:
systemically administering an effective amount of a 3-alkyl-4-aminobutyric acid or a 3-alkylglutamic acid and activating brain neuronal L-glutamic acid
decarboxylase activity.
14. A method as set forth in claim 13 wherein said administering step is further defined as administering a compound of the formula
Figure imgf000027_0001
wherein R1 is a straight chain or branched alkyl of 1 to 6 carbons, a phenyl or a cycloalkyl of 3 to 6 carbon atoms, R2 is -H, -CH3 or -CH3 and R3 is -H or -COOH, its diastereomers and enantiomers, and both pharamceutically acceptable base salts and acid addition salts thereof.
15. A method as set forth in claim 14 wherein the compound is 4-amino-3-(2-methylpropyl)butanoic acid.
16. A method as set forth in claim 14 wherein the compound is 4-amino-3-(2-methylpropyl) glutamic acid.
PCT/US1991/008701 1990-11-27 1991-11-20 Gaba and l-glutamic acid analogs for antiseizure treatment WO1992009560A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US61869290A 1990-11-27 1990-11-27
US618,692 1990-11-27

Publications (1)

Publication Number Publication Date
WO1992009560A1 true WO1992009560A1 (en) 1992-06-11

Family

ID=24478754

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1991/008701 WO1992009560A1 (en) 1990-11-27 1991-11-20 Gaba and l-glutamic acid analogs for antiseizure treatment

Country Status (3)

Country Link
AU (1) AU9137091A (en)
MX (1) MX9102241A (en)
WO (1) WO1992009560A1 (en)

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993023383A1 (en) * 1992-05-20 1993-11-25 Northwestern University Gaba and l-glutamic acid analogs for antiseizure treatment
WO1998003167A1 (en) 1996-07-24 1998-01-29 Warner-Lambert Company Isobutylgaba and its derivatives for the treatment of pain
WO1999031074A2 (en) * 1997-12-16 1999-06-24 Warner-Lambert Company ((cyclo)alkyl substituted)-.gamma.-aminobutyric acid derivatives (=gaba analogues), their preparation and their use in the treatment of neurological disorders
WO2000061234A1 (en) * 1999-04-09 2000-10-19 Warner-Lambert Company Combinations of gaba analogs and tricyclic compounds to treat depression
WO2000061135A1 (en) * 1999-04-08 2000-10-19 Warner-Lambert Company Method for the treatment of incontinence
US6153650A (en) * 1996-10-23 2000-11-28 Warner-Lambert Company Substituted gamma aminobutyric acids as pharmaceutical agents
WO2000076958A2 (en) * 1999-06-10 2000-12-21 Warner-Lambert Company Mono- and disubstituted 3-propyl gamma-aminobutyric acids
US6197819B1 (en) 1990-11-27 2001-03-06 Northwestern University Gamma amino butyric acid analogs and optical isomers
US6242488B1 (en) 1997-08-20 2001-06-05 University Of Oklahoma Method for preventing and treating pain
EP1192944A1 (en) * 2000-09-22 2002-04-03 Warner-Lambert Company Use of pregabalin for treating asthma
US6627771B1 (en) 1998-11-25 2003-09-30 Pfizer Inc Gamma amino butyric and acid analogs
US6642398B2 (en) 1999-06-10 2003-11-04 Warner-Lambert Company Mono-and disubstituted 3-propyl gamma-aminobutyric acids
WO2003104184A1 (en) 2002-06-11 2003-12-18 Xenoport, Inc. Methods for synthesis of acyloxyalkyl derivatives of gaba analogs
US6833140B2 (en) 2001-06-11 2004-12-21 Xenoport, Inc. Orally administered dosage forms of GABA analog prodrugs having reduced toxicity
WO2005025563A1 (en) * 2003-09-12 2005-03-24 Warner-Lambert Company Llc Combination comprising an alpha-2-delta ligand and an ssri and/or snri for treatment of depression and anxiety disorders
US6900192B2 (en) 2000-10-06 2005-05-31 Xenoport, Inc. Bile-acid conjugates for providing sustained systemic concentrations of drugs
US6927036B2 (en) 2002-02-19 2005-08-09 Xero Port, Inc. Methods for synthesis of prodrugs from 1-acyl-alkyl derivatives and compositions thereof
US6992109B1 (en) 1999-04-08 2006-01-31 Segal Catherine A Method for the treatment of incontinence
US6992076B2 (en) 2000-10-06 2006-01-31 Xenoport, Inc. Bile-acid derived compounds for providing sustained systemic concentrations of drugs after oral administration
US7026351B2 (en) 2002-03-20 2006-04-11 Xenoport, Inc. Cyclic 1-(acyloxy)-alkyl prodrugs of GABA analogs, compositions and uses thereof
US7060727B2 (en) 2002-12-11 2006-06-13 Xenoport, Inc. Prodrugs of fused GABA analogs, pharmaceutical compositions and uses thereof
US7183259B2 (en) 2002-05-17 2007-02-27 Xenoport Amino acid conjugates providing for sustained systemic concentrations of GABA analogues
US7186855B2 (en) 2001-06-11 2007-03-06 Xenoport, Inc. Prodrugs of GABA analogs, compositions and uses thereof
EP1820502A1 (en) 2006-02-10 2007-08-22 Laboratorios Del Dr. Esteve, S.A. Active substance combination comprising azolylcarbinol compounds
EP1840117A1 (en) * 1999-06-10 2007-10-03 Warner-Lambert Company LLC Mono- and disubstituted 3-propyl gamma-aminobutyric acids
US7420002B2 (en) 2001-06-11 2008-09-02 Xenoport Amino acid conjugates providing for sustained systemic concentrations of GABA analogues
WO2010042759A2 (en) 2008-10-08 2010-04-15 Kyphia Pharmaceuticals Inc Gaba conjugates and methods of use thereof
US7700652B2 (en) 2003-09-11 2010-04-20 Xenoport, Inc. Treating urinary incontinence using prodrugs of GABA analogs
WO2010099200A1 (en) * 2009-02-24 2010-09-02 Nektar Therapeutics Oligomer-amino acid conjugates
US7868043B2 (en) 2008-01-25 2011-01-11 Xenoport, Inc. Mesophasic forms of (3S)-aminomethyl-5-methyl-hexanoic acid prodrugs and methods of use
US7872046B2 (en) 2008-01-25 2011-01-18 Xenoport, Inc. Crystalline form of a (3S)-aminomethyl-5-methyl-hexanoic acid prodrug and methods of use
EP2343073A2 (en) 2003-12-11 2011-07-13 Sepracor Inc. Combination of a sedative and a neurotransmitter modulator, and methods for improving sleep quality and treating depression
US8026279B2 (en) 2003-10-14 2011-09-27 Xenoport, Inc. Crystalline form of γ-aminobutyric acid analog
US8048917B2 (en) 2005-04-06 2011-11-01 Xenoport, Inc. Prodrugs of GABA analogs, compositions and uses thereof
US8062870B2 (en) 2008-01-25 2011-11-22 Xenoport, Inc. Enantiomerically resolving acyloxyalkyl thiocarbonates used in synthesizing acyloxyalkyl carbamate prodrugs
US8114909B2 (en) 2003-09-17 2012-02-14 Xenoport, Inc. Treating or preventing restless legs syndrome using prodrugs of GABA analogs
WO2013060814A3 (en) * 2011-10-26 2013-06-20 Givaudan Sa Glutamic acid derivatives
WO2013121061A1 (en) 2012-02-15 2013-08-22 Laboratec, S.L. Pharmaceutical composition for treating urinary incontinence and enuresis
WO2014113299A1 (en) 2013-01-15 2014-07-24 Warsaw Orthopedic, Inc. Clonidine compounds in a biodegradable fiber
WO2014117176A1 (en) 2013-01-28 2014-07-31 Lopez Hector L Methods of improving tolerability, pharmacodynamics, and efficacy of b-alanine and use therefor
US8795725B2 (en) 2004-11-04 2014-08-05 Xenoport, Inc. GABA analog prodrug sustained release oral dosage forms
WO2015091505A1 (en) 2013-12-17 2015-06-25 Laboratorios Del Dr. Esteve, S.A. Gabapentinoids and sigma receptor ligands combinations
US9757358B2 (en) 2010-02-04 2017-09-12 Laboratorios Del Dr. Esteve, S.A. Sigma ligands for potentiating the analgesic effect of opioids and opiates in post-operative pain and attenuating the dependency thereof
US9782483B2 (en) 2010-05-21 2017-10-10 Laboratories Del Dr. Esteve, S.A. Sigma ligands for the prevention and/or treatment of emesis induced by chemotherapy or radiotherapy
US9789117B2 (en) 2011-05-18 2017-10-17 Laboratorios Del Dr. Esteve, S.A. Use of sigma ligands in diabetes type-2 associated pain
US9789115B2 (en) 2010-08-03 2017-10-17 Laboratorios Del Dr. Esteve, S.A. Use of sigma ligands in opioid-induced hyperalgesia
US9844516B2 (en) 2010-02-04 2017-12-19 Laboratorios De Dr. Esteve Sigma ligands for use in the prevention and/or treatment of post-operative pain
US9914705B2 (en) 2008-04-25 2018-03-13 Laboratorios Del Dr. Esteve, S.A. 1-aryl-3-aminoalkoxy pyrazoles as sigma ligands enhancing analgesic effect of opioids and attenuating the dependency thereof
US9931346B2 (en) 2013-12-17 2018-04-03 Laboratorios Del Dr. Esteve S.A. Serotonin-norepinephrine reuptake inhibitors (SNRIs) and Sigma receptor ligands combinations
DE202018105573U1 (en) 2018-09-27 2018-10-19 Laboratec Pharmaceutical composition for the treatment of urinary incontinence and enuresis
EP4227293A2 (en) 2018-05-14 2023-08-16 Xgene Pharmaceutical Inc Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalin

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2628980A (en) * 1949-02-18 1953-02-17 Gen Mills Inc Beta-phenyl-gamma-nitroaldehydes
JPS4940460B1 (en) * 1971-08-28 1974-11-02

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2628980A (en) * 1949-02-18 1953-02-17 Gen Mills Inc Beta-phenyl-gamma-nitroaldehydes
JPS4940460B1 (en) * 1971-08-28 1974-11-02

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
JOURNAL OF MEDICINAL CHEMISTRY, Volume 8, issued 1965, (Easton Pennsylvania), Y.C. KIM, "Glutamic Acid Analogs", see pages 509-513. *

Cited By (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6197819B1 (en) 1990-11-27 2001-03-06 Northwestern University Gamma amino butyric acid analogs and optical isomers
WO1993023383A1 (en) * 1992-05-20 1993-11-25 Northwestern University Gaba and l-glutamic acid analogs for antiseizure treatment
WO1998003167A1 (en) 1996-07-24 1998-01-29 Warner-Lambert Company Isobutylgaba and its derivatives for the treatment of pain
US6001876A (en) * 1996-07-24 1999-12-14 Warner-Lambert Company Isobutylgaba and its derivatives for the treatment of pain
USRE41920E1 (en) 1996-07-24 2010-11-09 Warner-Lambert Company Llc Isobutylgaba and its derivatives for the treatment of pain
US6153650A (en) * 1996-10-23 2000-11-28 Warner-Lambert Company Substituted gamma aminobutyric acids as pharmaceutical agents
US6242488B1 (en) 1997-08-20 2001-06-05 University Of Oklahoma Method for preventing and treating pain
US6426368B2 (en) 1997-08-20 2002-07-30 Warner-Lambert Company Method for preventing and treating alcoholism
WO1999031074A3 (en) * 1997-12-16 1999-11-04 Warner Lambert Co ((cyclo)alkyl substituted)-.gamma.-aminobutyric acid derivatives (=gaba analogues), their preparation and their use in the treatment of neurological disorders
EP1400526A2 (en) * 1997-12-16 2004-03-24 Warner-Lambert Company LLC ((cyclo)alkyl substituted)- gamma -aminobutyric acid derivatives (=Gaba analogues), their preparation and their use in the treatment of neurological disorders
EP1400526A3 (en) * 1997-12-16 2004-03-31 Warner-Lambert Company LLC ((cyclo)alkyl substituted)- gamma -aminobutyric acid derivatives (=Gaba analogues), their preparation and their use in the treatment of neurological disorders
WO1999031074A2 (en) * 1997-12-16 1999-06-24 Warner-Lambert Company ((cyclo)alkyl substituted)-.gamma.-aminobutyric acid derivatives (=gaba analogues), their preparation and their use in the treatment of neurological disorders
US6627771B1 (en) 1998-11-25 2003-09-30 Pfizer Inc Gamma amino butyric and acid analogs
WO2000061135A1 (en) * 1999-04-08 2000-10-19 Warner-Lambert Company Method for the treatment of incontinence
EP1977745A1 (en) * 1999-04-08 2008-10-08 Warner-Lambert Company LLC Method for the treatment of incontinence
US6992109B1 (en) 1999-04-08 2006-01-31 Segal Catherine A Method for the treatment of incontinence
WO2000061234A1 (en) * 1999-04-09 2000-10-19 Warner-Lambert Company Combinations of gaba analogs and tricyclic compounds to treat depression
US6642398B2 (en) 1999-06-10 2003-11-04 Warner-Lambert Company Mono-and disubstituted 3-propyl gamma-aminobutyric acids
WO2000076958A3 (en) * 1999-06-10 2001-04-12 Warner Lambert Co Mono- and disubstituted 3-propyl gamma-aminobutyric acids
WO2000076958A2 (en) * 1999-06-10 2000-12-21 Warner-Lambert Company Mono- and disubstituted 3-propyl gamma-aminobutyric acids
KR100694735B1 (en) * 1999-06-10 2007-03-14 워너-램버트 캄파니 엘엘씨 Mono- and Disubstituted 3-Propyl Gamma-Aminobutyric Acids
CZ301608B6 (en) * 1999-06-10 2010-04-28 Warner-Lambert Company 3-Propyl gamma aminobutyric acid derivative and pharmaceutical composition in which the derivative is comprised
AP1397A (en) * 1999-06-10 2005-04-19 Warner Lambert Co Mono-and disubstituted 3-propyl gamma-aminobutyric acids.
CZ300834B6 (en) * 1999-06-10 2009-08-19 Warner-Lambert Company Llc Mono- and disubstituted 3-propyl gamma-aminobutyric acids
EP1840117A1 (en) * 1999-06-10 2007-10-03 Warner-Lambert Company LLC Mono- and disubstituted 3-propyl gamma-aminobutyric acids
EP1192944A1 (en) * 2000-09-22 2002-04-03 Warner-Lambert Company Use of pregabalin for treating asthma
US6992076B2 (en) 2000-10-06 2006-01-31 Xenoport, Inc. Bile-acid derived compounds for providing sustained systemic concentrations of drugs after oral administration
US6900192B2 (en) 2000-10-06 2005-05-31 Xenoport, Inc. Bile-acid conjugates for providing sustained systemic concentrations of drugs
US7049305B2 (en) 2000-10-06 2006-05-23 Xenoport, Inc. Bile-acid conjugates providing for sustained systemic concentration of drugs
US7601706B2 (en) 2000-10-06 2009-10-13 Xenoport, Inc. Bile-acid conjugates providing for sustained systemic concentration of drugs
US7601708B2 (en) 2000-10-06 2009-10-13 Xenoport, Inc. Bile-acid derived compounds for providing sustained systemic concentrations of drugs after oral administration
EP2085087A1 (en) 2001-06-11 2009-08-05 Xenoport, Inc. Prodrugs of gaba analogs, compositions and uses thereof
US7790708B2 (en) 2001-06-11 2010-09-07 Xenoport, Inc. Prodrugs of GABA analogs, compositions and uses thereof
US8168623B2 (en) 2001-06-11 2012-05-01 Xenoport, Inc. Prodrugs of GABA analogs, compositions and uses thereof
US6972341B2 (en) 2001-06-11 2005-12-06 Xeno Port, Inc. Prodrugs of GABA analogs, compositions and uses thereof
US7420002B2 (en) 2001-06-11 2008-09-02 Xenoport Amino acid conjugates providing for sustained systemic concentrations of GABA analogues
US6833140B2 (en) 2001-06-11 2004-12-21 Xenoport, Inc. Orally administered dosage forms of GABA analog prodrugs having reduced toxicity
US9238616B2 (en) 2001-06-11 2016-01-19 Xenoport, Inc. Prodrugs of gaba analogs, compositions and uses thereof
US7645797B2 (en) 2001-06-11 2010-01-12 Xenoport, Inc. Amino acid conjugates providing for sustained systemic concentrations of GABA analogues
EP1902714B1 (en) * 2001-06-11 2012-01-11 XenoPort, Inc. Prodrugs of gaba analogs, compositions and uses thereof
US7186855B2 (en) 2001-06-11 2007-03-06 Xenoport, Inc. Prodrugs of GABA analogs, compositions and uses thereof
US8143437B2 (en) 2002-02-19 2012-03-27 Xenoport, Inc. Methods for synthesis of prodrugs from 1-acyl-alkyl derivatives and compositions thereof
US7560483B2 (en) 2002-02-19 2009-07-14 Xenoport, Inc. Methods for synthesis of prodrugs from 1-acyl-alkyl derivatives and compositions thereof
US6927036B2 (en) 2002-02-19 2005-08-09 Xero Port, Inc. Methods for synthesis of prodrugs from 1-acyl-alkyl derivatives and compositions thereof
US7868041B2 (en) 2002-03-20 2011-01-11 Xenoport, Inc. Cyclic 1-(acyloxy)-alkyl prodrugs of GABA analogs, compositions and uses thereof
US7026351B2 (en) 2002-03-20 2006-04-11 Xenoport, Inc. Cyclic 1-(acyloxy)-alkyl prodrugs of GABA analogs, compositions and uses thereof
US7569576B2 (en) 2002-03-20 2009-08-04 Xenoport, Inc. Cyclic 1-(acyloxy)-alkyl prodrugs of GABA analogs, compositions and uses thereof
US7767649B2 (en) 2002-05-17 2010-08-03 Xenoport, Inc. Amino acid conjugates providing for sustained systemic concentrations of GABA analogues
US7183259B2 (en) 2002-05-17 2007-02-27 Xenoport Amino acid conjugates providing for sustained systemic concentrations of GABA analogues
WO2003104184A1 (en) 2002-06-11 2003-12-18 Xenoport, Inc. Methods for synthesis of acyloxyalkyl derivatives of gaba analogs
EP2275401A1 (en) 2002-06-11 2011-01-19 XenoPort, Inc. Crystalline 1-{[(a-isobutanoyloxyethoxy)carbonyl]aminomethyl}-1-cyclohexane acetic acid
US7060727B2 (en) 2002-12-11 2006-06-13 Xenoport, Inc. Prodrugs of fused GABA analogs, pharmaceutical compositions and uses thereof
US7700652B2 (en) 2003-09-11 2010-04-20 Xenoport, Inc. Treating urinary incontinence using prodrugs of GABA analogs
WO2005025563A1 (en) * 2003-09-12 2005-03-24 Warner-Lambert Company Llc Combination comprising an alpha-2-delta ligand and an ssri and/or snri for treatment of depression and anxiety disorders
US8114909B2 (en) 2003-09-17 2012-02-14 Xenoport, Inc. Treating or preventing restless legs syndrome using prodrugs of GABA analogs
US8686034B2 (en) 2003-10-14 2014-04-01 Xenoport, Inc. Crystalline form of γ-aminobutyric acid analog
US8026279B2 (en) 2003-10-14 2011-09-27 Xenoport, Inc. Crystalline form of γ-aminobutyric acid analog
US9150503B2 (en) 2003-10-14 2015-10-06 Xenoport, Inc. Crystalline form of γ-aminobutyric acid analog
EP2343073A2 (en) 2003-12-11 2011-07-13 Sepracor Inc. Combination of a sedative and a neurotransmitter modulator, and methods for improving sleep quality and treating depression
US8906412B2 (en) 2004-11-04 2014-12-09 Xenoport, Inc. GABA analog prodrug sustained release oral dosage forms
US8795725B2 (en) 2004-11-04 2014-08-05 Xenoport, Inc. GABA analog prodrug sustained release oral dosage forms
US8048917B2 (en) 2005-04-06 2011-11-01 Xenoport, Inc. Prodrugs of GABA analogs, compositions and uses thereof
EP1820502A1 (en) 2006-02-10 2007-08-22 Laboratorios Del Dr. Esteve, S.A. Active substance combination comprising azolylcarbinol compounds
US7872046B2 (en) 2008-01-25 2011-01-18 Xenoport, Inc. Crystalline form of a (3S)-aminomethyl-5-methyl-hexanoic acid prodrug and methods of use
US7868043B2 (en) 2008-01-25 2011-01-11 Xenoport, Inc. Mesophasic forms of (3S)-aminomethyl-5-methyl-hexanoic acid prodrugs and methods of use
US8062870B2 (en) 2008-01-25 2011-11-22 Xenoport, Inc. Enantiomerically resolving acyloxyalkyl thiocarbonates used in synthesizing acyloxyalkyl carbamate prodrugs
US8258179B2 (en) 2008-01-25 2012-09-04 Xenoport, Inc. Crystalline form of a (3S)-aminomethyl-5-methyl-hexanoic acid prodrug and methods of use
US9914705B2 (en) 2008-04-25 2018-03-13 Laboratorios Del Dr. Esteve, S.A. 1-aryl-3-aminoalkoxy pyrazoles as sigma ligands enhancing analgesic effect of opioids and attenuating the dependency thereof
EP3075722A1 (en) 2008-10-08 2016-10-05 Xgene Pharmaceutical Inc Gaba conjugates and methods of use thereof
WO2010042759A2 (en) 2008-10-08 2010-04-15 Kyphia Pharmaceuticals Inc Gaba conjugates and methods of use thereof
US10478412B2 (en) 2008-10-08 2019-11-19 Xgene Pharmaceutical Inc. GABA conjugates and methods of use thereof
US9186341B2 (en) 2008-10-08 2015-11-17 Feng Xu GABA conjugates and methods of use thereof
US8268887B2 (en) 2008-10-08 2012-09-18 Feng Xu Drug conjugates and methods of use thereof
WO2010099200A1 (en) * 2009-02-24 2010-09-02 Nektar Therapeutics Oligomer-amino acid conjugates
US9192681B2 (en) 2009-02-24 2015-11-24 Nektar Therapeutics Oligomer-amino acid conjugates
US9487473B2 (en) 2009-02-24 2016-11-08 Nektar Therapeutics Oligomer-amino acid conjugates
US9844516B2 (en) 2010-02-04 2017-12-19 Laboratorios De Dr. Esteve Sigma ligands for use in the prevention and/or treatment of post-operative pain
US9757358B2 (en) 2010-02-04 2017-09-12 Laboratorios Del Dr. Esteve, S.A. Sigma ligands for potentiating the analgesic effect of opioids and opiates in post-operative pain and attenuating the dependency thereof
US9782483B2 (en) 2010-05-21 2017-10-10 Laboratories Del Dr. Esteve, S.A. Sigma ligands for the prevention and/or treatment of emesis induced by chemotherapy or radiotherapy
US9789115B2 (en) 2010-08-03 2017-10-17 Laboratorios Del Dr. Esteve, S.A. Use of sigma ligands in opioid-induced hyperalgesia
US9789117B2 (en) 2011-05-18 2017-10-17 Laboratorios Del Dr. Esteve, S.A. Use of sigma ligands in diabetes type-2 associated pain
WO2013060814A3 (en) * 2011-10-26 2013-06-20 Givaudan Sa Glutamic acid derivatives
US9272011B2 (en) 2012-02-15 2016-03-01 Laboratec, S.L. Pharmaceutical composition for treating urinary incontinence and enuresis
WO2013121061A1 (en) 2012-02-15 2013-08-22 Laboratec, S.L. Pharmaceutical composition for treating urinary incontinence and enuresis
WO2014113299A1 (en) 2013-01-15 2014-07-24 Warsaw Orthopedic, Inc. Clonidine compounds in a biodegradable fiber
WO2014117176A1 (en) 2013-01-28 2014-07-31 Lopez Hector L Methods of improving tolerability, pharmacodynamics, and efficacy of b-alanine and use therefor
WO2015091505A1 (en) 2013-12-17 2015-06-25 Laboratorios Del Dr. Esteve, S.A. Gabapentinoids and sigma receptor ligands combinations
US9931346B2 (en) 2013-12-17 2018-04-03 Laboratorios Del Dr. Esteve S.A. Serotonin-norepinephrine reuptake inhibitors (SNRIs) and Sigma receptor ligands combinations
EP4227293A2 (en) 2018-05-14 2023-08-16 Xgene Pharmaceutical Inc Crystalline forms of 1-(acyloxy)-alkyl carbamate drug conjugates of naproxen and pregabalin
DE202018105573U1 (en) 2018-09-27 2018-10-19 Laboratec Pharmaceutical composition for the treatment of urinary incontinence and enuresis

Also Published As

Publication number Publication date
AU9137091A (en) 1992-06-25
MX9102241A (en) 1994-01-31

Similar Documents

Publication Publication Date Title
WO1992009560A1 (en) Gaba and l-glutamic acid analogs for antiseizure treatment
Elliott et al. Gamma-aminobutyric acid
EP0804183B1 (en) Use of creatine or creatine analogs for the treatment of huntigton's disease, parkinson's disease and amyotrophic lateral sclerosis
EP0934061B1 (en) Isobutylgaba and its derivatives for the treatment of pain
Palfreyman et al. The pharmacology of GABA-transaminase inhibitors
CN101199501A (en) HIF hydroxylase inhibitors
Baran et al. Effect of mannitol treatment on brain neurotransmitter markers in kainic acid-induced epilepsy
US4271192A (en) Process for treatment and prevention of ventricular fibrillation
CA2324426A1 (en) Compositions and methods for the treatment of cystic fibrosis
PL213698B1 (en) Treatment regimen for administration of phenylacetylglutamine, phenylacetylisoglutamine, and/or phenylacetate
Seiler Ornithine aminotransferase, a potential target for the treatment of hyperammonemias
Carvajal et al. Anticonvulsive action of substances designed as inhibitors of γ-aminobutyric acid-α-ketoglutaric acid transaminase
EP0124091B1 (en) Treatment of seizure disorders and pharmaceutical compositions useful therein
US4389415A (en) Method of treating hypertension
US20100062987A1 (en) Anticonvulsive pharmaceutical compositions
EP0637449B1 (en) Use of L-carnitine or acyl L-carnitines and valproate for treating seizure disorders
JP2003516962A (en) Adenosine kinase inhibitors for the treatment of optic nerve and retinal damage
Seiler et al. Amplification by glycine of the anticonvulsant effect of THPO, a GABA uptake inhibitor
CA2083507A1 (en) Nicotinylalanine as a therapeutic agent acting on the central nervous system
Bernstein et al. Short-circuiting the visual cycle with retinotoxic aromatic amines.
EP0003353A1 (en) A pharmaceutical composition for oral administration for treating hypertension
Endou et al. Ammonia Metabolism in Human Kidney
MXPA04005145A (en) Use of anti-glaucoma drugs to treat visual defects associated with the use of a gabaergic agent.
AU2008207610A1 (en) Method for modulating macrophage activation
AU6246600A (en) Use of creatine analogues for the treatment of disorders of glucose metabolism

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA FI JP KR NO

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LU NL SE

NENP Non-entry into the national phase

Ref country code: CA