Isobutylgaba and its derivatives for the treatment of pain

The instant invention is a method of using certain analogs of glutamic acid and gamma-aminobutyric acid in pain therapy.

Skip to: Description  ·  Claims  ·  References Cited  · Patent History  ·  Patent History
Description

Notice: more than one reissue application has been filed for the reissue of U.S. Pat. No. 6,001,876. The reissue applications: are U.S. application Ser. No. 12/700,968 (filed Feb. 5, 2010), which is a continuation of the present application; and, the present application.

This application claims benefit of Provisional application Ser. No. 60/022,337, Jul. 24, 1996.

BACKGROUND OF THE INVENTION

The present invention is the use of analogs of glutamic acid and gamma-aminobutyric acid (GABA) in pain therapy, as the compounds exhibit analgesic/antihyperalgesic action. Advantages of the use of the compounds includes the finding that repeated use does not lead to tolerance nor is there a cross-tolerance between morphine and the compounds.

The compounds of the invention are known agents useful in antiseizure therapy for central nervous system disorders such as epilepsy, Huntington's chorea, cerebral ischemia, Parkinson's disease, tardive dyskinesia, and spasticity. It has also been suggested that the compounds can be used as antidepressants, anxiolytics, and antipsychotics. See WO 92/09560 (U.S. Ser. No. 618,692 filed Nov. 27, 1990) and WP 93/23383 (U.S. Ser. No. 886,080 filed May 20, 1992).

SUMMARY OF THE INVENTION

The instant invention is a method of using a compound of Formula I below in the treatment of pain, especially for treatment of chronic pain disorders. Such disorders include, but are not limited to, inflammatory pain, postoperative pain, osteoarthritis pain associated with metastatic cancer, trigeminal neuralgia, acute herpetic and postherpetic neuralgia, diabetic neuropathy, causalgia, brachial plexus avulsion, occipital neuralgia, reflex sympathetic dystrophy, fibromyalgia, gout, phantom limb pain, bum burn pain, and other forms of neuralgic, neuropathic, and idiopathic pain syndromes.

A compound are those of Formula I
or a pharmaceutically acceptable salt thereof wherein

    • R1 is a straight or branched alkyl of from 1 to 6 carbon atoms, phenyl, or cycloalkyl of from 3 to 6 carbon atoms;
    • R2 is hydrogen or methyl; and
    • R3 is hydrogen, methyl, or carboxyl.

Diastereomers and enantiomers of compounds of Formula I are included in the invention.

Preferred compounds of the invention are those according to claim 1 wherein R3 and R2 are hydrogen, and R1 is —(CH2)0-2—i C4H9 as an (R), (S), or (R,S) isomer.

The more preferred compounds of the invention are (S)-3-(aminomethyl)-5-methylhexanoic acid and 3-aminomethyl-5-methyl-hexanoic acid.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1. Effect of Gabapentin (1-(aminomethyl)-cyclohexaneacetic acid), CI-1008 ((S)-3-(aminomethyl)-5-methylhexanoic acid), and 3-aminomethyl-5-methylhexanoic acid in the Rat Paw Formalin Test

Test compounds were administered s.c. 1 hour before an intraplantar injection of 50 μL formalin. The time spent licking/biting the injected paw during the early and late phases was scored. Results are shown as the mean ±SEM of 6 to 8 animals per group. *P<0.05 and **P<0.01 significantly different from vehicle (Veh.) treated controls (ANOVA followed by Dunnett's t-test).

FIG. 2. Effect of Gabapentin and CI-1008 on Carrageenin-Induced Mechanical Hyperalgesia

Nociceptive pressure thresholds were measured in the rat using the paw pressure test. Baseline (BL) measurements were taken before animals were administered with 100 μL of 2% carrageenin by intraplantar injection. Results are shown as mean (±SEM) of 8 animals per group. Gabapentin (GP), CI-1008, or morphine (MOR; 3 mg/g) was administered s.c. 3.5 hours after carrageenin. *P<0.05 and **P<0.01 significantly different from vehicle control group at the same time point (ANOVA followed by Dunnett's t-test).

FIG. 3. Effect of Gabapentin and CI-1008 on Carrageenin-Induced Thermal Hyperalgesia

Nociceptive thermal thresholds were measured in the rat using the Hargreaves apparatus. Baseline (BL) measurements were taken before animal s were administered with 100 μL of 2% carrageenin by intraplantar injection. Results are shown as mean (±SEM) of 8 animals per group. Gabapentin (GP) or CI-1008 was administered s.c. 2.5 hours after carrageenin. *P<0.05 and **P<0.01 significantly different from vehicle control group at the same time point (ANOVA followed by Dunnett's t-test).

FIG. 4. Effect of (a) Morphine, (b) Gabapentin, and (c) S-(+)-3-Isobutylgaba on Thermal Hyperalgesia in the Rat Postoperative Pain Model

Gabapentin or S-(+)-3 isobutylgaba was administered 1 hour before surgery. Morphine was administered 0.5 hour before surgery. Thermal paw withdrawal latencies (PWL) were determined for both ipsilateral and contralateral paws using the rat plantar test. For clarity contralateral paw data for drug-treated animals is not shown. Baseline (BL) measurements were taken before surgery and PWL were reassessed 2, 24, 48, and 72 hours postsurgery. Results are expressed as the mean PWL(s) of 8 to 10 animals per group (vertical bars represent ±SEM). *P<0.05 **P<0.01 significantly different (ANOVA followed by Dunnett's t-test), comparing ipsilateral paw of drug-treated groups to ispsilateral paw of vehicle-treated group at each time point. In the figure, —●— is vehicle contralateral, —◯— is vehicle ispsilateral, —Δ— is 1 mg/kg morphine, —▭— is 3, and —⋄— is 6 for morphine in 4a. In 4b, —Δ— is 3, —▭— is 10, and —⋄— is 30 for gabapentin. In 4c, —Δ— is 3 mg/kg, —▭— is 10, and —⋄— is 30 for S-(+)-isobutylgaba.

FIG. 5 Effect of (a) Morphine, (b) Gabapentin, and (c) S-(+)-3-Isobutylgaba on Tactile Allodynia in the Rat Post-operative Pain Model

Gabapentin or S-(+)-3-isobutylgaba was administered 1 hour before surgery. Morphine was administered 0.5 hour before surgery. Paw withdrawal thresholds to von Frey hair filaments were determined for both ipsilateral and contralateral paws. For clarity, contralateral paw data for drug-treated animals is not shown. Baseline (BL) measurements were taken before surgery, and withdrawal thresholds were reassessed 3, 25, 49, and 73 hours postsurgery. Results are expressed as median force (g) required to induce a withdrawal of paw in 8 to 10 animals per group (vertical bars represent first and third quartiles). *P<0.05 significantly different (Mann Whitney t-test) comparing ipsilateral paw of drug-treated groups to ipsilateral paw of vehicle treated group at each time point. In FIG. 5, —●— is vehicle contralateral, —◯— is vehicle ispsilateral. For morphine (5a), —Δ— is 1 mg/kg, —▭— is 3, and —⋄— is 16.

In 5b for gabapentin and S-(+)-isobutylgaba, —Δ— is 3 mg/kg, —▭— is 10, and —⋄— is 30.

FIG. 6. Effect of S-(+)-3-Isobulylgaba on the Maintenance of (a) Thermal Hyperalgesia and (b) Tactile Allodynia in the Rat Postoperative Pain Model.

S-(+)-3-Isobutylgaba (S-(+)-IBG) was administered 1 hour after surgery. Thermal paw withdrawal latencies, determined using the rat plantar test, and paw withdrawal thresholds to von Frey hair filaments, were determined in separate groups of animals for both ipsilateral and contralateral paws. For clarity only the ipsilateral paw data is shown. Baseline (BL) measurements were taken before surgery and withdrawal thresholds were reassessed up to 6 hours postsurgery. For thermal hyperalgesia, the results are expressed as the mean PWL(s) of 6 animals per group (vertical bars represent ±SEM), *P<0.05 **P<0.01 significantly different (unpaired t-test), comparing ipsilateral paw of drug-treated group to ipsilateral paw of vehicle (Veh —◯—) treated group at each time point. For tactile allodynia, the results are expressed as median force (g) required to induce a paw withdrawal of 6 animals per group (vertical bars represent first and third quartiles). *P<0.05 significantly different (Mann Whitney t-test), comparing ipsilateral paw of drug-treated group to ipsilateral paw of vehicle-treated group at each time point. —●— is S-(+)-IBG at 30 mg/kg.

DETAILED DESCRIPTION

The instant invention is a method of using a compound of Formula I above as an analgesic in the treatment of pain as listed above. Pain such as inflammatory pain, neuropathic pain, cancer pain, postoperative pain, and idiopathic pain which is pain of unknown origin, for example, phantom limb pain are included especially. Neuropathic pain is caused by injury or infection of peripheral sensory nerves. It includes, but is not limited to pain from peripheral nerve trauma, herpes virus infection, diabetes mellitus, causalgia, plexus avulsion, neuroma, limb amputation, and vasculitis. Neuropathic pain is also caused by nerve damage from chronic alcoholism, human immunodeficiency virus infection, hypothyroidism, uremia, or vitamin deficiencies. Neuropathic pain includes, but is not limited to pain caused by nerve injury such as, for example, the pain diabetics suffer from.

The conditions listed above are known to be poorly treated by currently marketed analgesics such as narcotics or nonsteroidal anti-inflammatory drugs (NSAID) due to insufficient efficacy or limiting side effects.

The terms used in Formula I are, for example, alkyl which term is methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, isopentyl, and neopentyl, as well as those as would occur to one skilled in the art.

The term “cycloalkyl” is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.

The compounds of 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 of 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 (Kim Y. C., Cocolase G. H., J. Med. Chem., 1965:8509), 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 β-hydroxyester with phosphoryl chloride and pyridine. The Michael addition of nitromethane to α,β-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 catalytic 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 quantitatively 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., Tetrahedrom. Lett., 1975:2647-2650.

The compounds made by the synthetic methods can be used as pharmaceutical compositions as agent in the treatment of pain when an effective amount of a compound of the Formula I, together with a pharmaceutically acceptable carrier is used. 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 by either oral or parenteral routes of administration. For example, these pharmaceutical compositions can be made in inert, pharmaceutically acceptable carriers which are either solid or liquid. Solid form preparations 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 and administered by the oral route in an appropriate formulation, or by a parenteral route such as intravenous, intramuscular, or subcutaneous injection as a liquid formulation.

The quantity of active compound in a unit dose of preparation may be varied or adjusted from 1 mg to about 300 mg/kg daily, based on an average 70-kg patient. A daily dose range of about 1 mg to about 50 mg/kg 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.

Effects of Gabapentin, CI-1008, and 3-Aminomethyl-5-methyl-hexanoic Acid in the Rat Formalin Paw Test

Male Sprague-Dawley rats (70-90 g) were habituated to perspex observation chambers (24 cm×24 cm×24 cm) for at least 15 minutes prior to testing. Formalin-induced hind paw licking and biting was initiated by a 50 μL subcutaneous injection of a 5% formalin solution (5% formaldehyde in isotonic saline) into the plantar surface of the left hind paw. Immediately following the formalin injection, licking/biting of the injected hind paw was scored in 5 minute bins for 60 minutes. The results are expressed as mean combined licking/biting time for the early phase (0-10 minutes) and late phase (10-45 minutes).

The s.c. administration of gabapentin (10-300 mg/kg) or CI-1008 (1-100 mg/kg) 1 hour before formalin dose-dependently blocked the licking/biting behavior during the late phase of the formalin response with respective minimum effective doses (MED) of 30 and 10 mg/kg (FIG. 1). However, neither of the compounds affected the early phase at any of the doses tested. Similar administration of 3-aminomethyl-5-methyl-hexanoic acid produced only a modest blockade of the late phase at 100 mg/kg.

Effects of Gabapentin and CI-1008 on Carrageenin-Induced Hyperalgesia

On the test Day, 2 to 3 baseline measurements were taken before rats (male Sprague-Dawley 70-90 g) were administered with 100 μL of 2% carrageenin by intraplantar injection into the right hind paw. Animals were dosed with the test drug after development of peak hyperalgesia. Separate groups of animals were used for the mechanical and thermal hyperalgesia studies.

A. Mechanical Hyperalgesia

Nociceptive pressure thresholds were measured in the rat paw pressure test using an analgesimeter (Ugo Basile). A cut-off point of 250 g was used to prevent any damage to the paw. The intraplantar injection of carrageenin produced a reduction in the nociceptive pressure threshold between 3 and 5 hours after injection, indicating induction of hyperalgesia. Morphine (3 mg/kg, s.c.) produced a complete blockade of hyperalgesia (FIG. 2). Gabapentin (3-300 mg/kg, s.c.) and CI-1008 (1-100 mg/kg, s.c.) dose-dependently antagonized the hyperalgesia, with respective MED of 10 and 3 mg/kg (FIG. 2).

B. Thermal Hyperalgesia

Baseline paw withdrawal latencies (PWL) were obtained for each rat using the Hargreaves model. Carrageenin was injected as described above. Animals were then tested for thermal hyperalgesia at 2 hours postcarrageenin administration. Gabapentin (10-100 mg/kg) or CI-1008 (1-30 mg/kg) was administered s.c. 2.5 hours after carrageenin, and PWL were reevaluated at 3 and 4 hours postcarrageenin administration. Carrageenin induced a significant reduction in paw withdrawal latency at 2, 3, and 4 hours following injection, indicating the induction of thermal hyperalgesia (FIG. 3). Gabapentin and CI-1008 dose-dependently antagonized the hyperalgesia with a MED of 30 and 3 mg/kg (FIG. 3).

These data show that gabapentin and CI-1008 are effective in the treatment of inflammatory pain.

The assay of Bennett G. J. provides an animal model of a peripheral mononeuropathy in rat that produces disorder of pain sensation like those seen in man (Pain, 1988;33:87-107).

The assay of Kim S. H., et al., provides one experimental model for peripheral neuropathy produced by segmented spinal nerve ligation in the rat (Pain, 1990;50:355-363).

A rat model of postoperative pain has been described (Brennan et al., 1996). It involves an incision of the skin, fascia, and muscle of the plantar aspect of the hind paw. This leads to an induction of reproducible and quantifiable mechanical hyperalgesia lasting several days. It has been suggested that this model displays some similarities to the human postoperative pain state. In the present study we have examined and compared the activities of gabapentin and S-(+)-3-isobutylgaba with morphine in this model of post-operative pain.

METHODS

Male Sprague-Dawley rats (250-300 g), obtained from Bantin and Kingmen, (Hull, U. K.) were used in all experiments. Before surgery, animals were housed in groups of 6 under a 12-hour light/dark cycle (lights on at 07 hour 00 minute) with food and water ad libitum. Postoperatively, animals were housed in pairs on “Aqua-sorb” bedding consisting of air laid cellulose (Beta Medical and Scientific, Sale, U.K.) under the same conditions. All experiments were carried out by an observer blind to drug treatments.

Surgery

Animals were anaesthetized with 2% isofluorane and 1.4 O2/NO2 mixture which was maintained during surgery via a nose cone. The plantar surface of the right hind paw was prepared with 50% ethanol, and a 1-cm longitudinal incision was made through skin and fascia, starting 0.5 cm from the edge of the heel and extending towards the toes. The plantaris muscle was elevated using forceps and incised longitudinally. The wound was closed using two simple sutures of braided silk with a FST-02 needle. The wound site was covered with Terramycin spray and Auromycin powder. Postoperatively, none of the animals displayed any signs of infection with the wounds healing well after 24 hours. The sutures were removed after 48 hours.

Evaluation of Thermal Hyperalgesia

Thermal hyperalgesia was assessed using the rat plantar test (Ugo Basile, Italy) following a modified method of Hargreaves, et al., 1988. Rats were habituated to the apparatus which consisted of three individual perspex boxes on an elevated glass table. A mobile radiant heat source was located under the table and focused onto the hind paw and paw withdrawal latencies (PWL) were recorded. There was an automatic cut off point of 22.5 seconds to prevent tissue damage. PWLs were taken 2 to 3 times for both hind paws of each animal, the mean of which represented baselines for right and left hind paws. The apparatus was calibrated to give a PWL of approximately 10 seconds. PWL(s) were reassessed following the same protocol as above 2, 24, 48, and 72 hours postoperatively.

Evaluation of Tactile Allodynia

Tactile allodynia was measured using Semmes-Weinstein von Frey hairs (Stoelting, Ill., U.S.A.). Animals were placed into wire-mesh-bottom cages allowing access to the underside of their paws. The animals were habituated to this environment prior to the start of the experiment. Tactile allodynia was tested by touching the plantar surface of the animals hind paw with von Frey hairs in ascending order of force (0.7, 1.2, 1.5, 2, 3.6, 5.5, 8.5, 11.8, 15. 1, and 29 g) until a paw withdrawal response was elicited. Each von Frey hair was applied to the paw for 6 seconds, or until a response occurred. Once a withdrawal response was established, the paw was retested, starting with the next descending von Frey hair until no response occurred. The highest force of 29 g lifted the paw as well as eliciting a response, thus represented the cut-off point. Each animal had both hind paws tested in this manner. The lowest amount of force required to elicit a response was recorded as withdrawal threshold in grams. When compounds were administered before surgery, the same animals were used to study drug effects on tactile, allodynia, and thermal hyperalgasia, with each animal being tested for tactile allodynia 1 hour after thermal hyperalgesia. Separate groups of animals were used for examination of tactile allodynia and thermal hyperalgesia when S-(+)-3-isobutylgaba was administered after surgery.

Statistics

Data obtained for thermal hyperalgesia was subjected to a one-way (analysis of variance) ANOVA followed by a Dunnett's t-test. Tactile allodynia results obtained with the von Frey hairs were subjected to an individual Mann Whitney t-test.

RESULTS

An incision of the rat plantaris muscle led to an induction of thermal hyperalgesia and tactile allodynia. Both nociceptive responses peaked within 1 hour following surgery and were maintained for 3 days. During the experimental period, all animals remained in good health.

Effect of Gabapentin. S-(+)-3-Isobutylgaba and Morphine Administered Before Surgery on Thermal Hyperalgesia

The single-dose administration of gabapentin 1 hour before surgery dose-dependently (3-30 mg/kg, s.c.) blocked development of thermal hyperalgesia with a MED of 30 mg/kg (FIG. 1b). The highest dose of 30 mg/kg gabapentin prevented the hyperalgesic response for 24 hours (FIG. 1b). Similar administration of S-(+)-3-isobutylgaba also dose-dependently (3-30 mg/kg, s.c.) prevented development of thermal hyperalgesia with a MED of 3 mg/kg (FIG. 1c). The 30 mg/kg dose of S-(+)-3-isobutylgaba was effective up to 3 days (FIG. 1c). The administration of morphine 0.5 hour before surgery dose-dependently (1-6 mg/kg, s.c.) antagonized the development of thermal hyperalgesia with a MED of 1 mg/kg (FIG. 1a). This effect was maintained for 24 hours (FIG. 1a).

Effects of Gabapentin, S-(+)-3-Isobutylgaba and Morphine Administered Before Surgery on Tactile Allodynia

The effect of drugs on development of tactile allodynia was determined in the same animals used for thermal hyperalgesia above. One hour was allowed between thermal hyperalgesia and tactile allodynia tests. Gabapentin dose-dependently prevented development of tactile allodynia with a MED of 10 mg/kg. The 10 and 30 mg/kg doses of gabapentin were effective for 25 and 49 hours, respectively (FIG. 2b). S-(+)-3-Isobutylgaba also dose-dependently (3-30 mg/kg) blocked development of the allodynia response with a MED of 10 mg/kg (FIG. 2c). This blockade of the nociceptive response was maintained for 3 days by the 30 mg/kg dose of S-(+)-3-isobutylgaba (FIG. 2c). In contrast, morphine (1-6 mg/kg) only prevented the development of tactile allodynia for 3 hour postsurgery at the highest dose of 6 mg/kg (FIG. 2a).

Effect of S-(+)-3-Isobutylgaba Administered 1 Hour After Surgery on Tactile Allodynia and Thermal Hyperalgesia

The allodynia and hyperalgesia peaked within 1 hour in all animals and was maintained for the following 5 to 6 hours. The s.c. administration of 30 mg/kg S-(+)-3-isobutylgaba 1 hour after surgery blocked the maintenance of tactile allodynia and thermal hyperalgesia for 3 to 4 hours. After this time, both nociceptive responses returned to control levels indicating disappearance of antihyperalgesic and antiallodynic actions (FIG. 3).

Gabapentin and S-(+)-3-isobutylgaba did not affect PWL in the thermal hyperalgesia test or tactile allodynia scores in the contralateral paw up to the highest dose tested in any of the experiments. In contrast, morphine (6 mg, s.c.) increased PWL of the contralateral paw in the thermal hyperalgesia test (data not shown).

The results presented here show that incision of the rat plantaris muscle induces thermal hyperalgesia and tactile allodynia lasting at least 3 days. The major findings of the present study are that gabapentin and S-(+)-3-isobutylgaba are equally effective at blocking both nociceptive responses. In contrast, morphine was found to be more effective against thermal hyperalgesia than tactile allodynia. Furthermore, S-(+)-3-isobutylgaba completely blocked induction and maintenance of allodynia and hyperalgesia.

Claims

1. A method for treating pain comprising administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt, diastereomer, or enantiomer thereof wherein

R1 is a straight or branched alkyl of from 1 to 6 carbon atoms, phenyl, or cycloalkyl of from 3 to 6 carbon atoms;
R2 is hydrogen or methyl; and
R3 is hydrogen, methyl, or carboxyl to a mammal in need of said treatment.

2. A method according to claim 1 for treating pain comprising administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof, wherein the compound administered is a compound of Formula I wherein R3 and R2 are hydrogen, and R1 is —(CH2)0-2—i C4H9 isobutyl as an (R), (S), or (R,S) isomer, to a mammal in need of said treatment.

3. A method according to claim 1 wherein the compound administered is named (S)-3-(aminomethyl)-5-methylhexanoic acid and 3-aminomethyl-5-methylhexanoic acid.

4. A method according to claim 1 2 wherein the pain treated is inflammatory pain.

5. A method according to claim 1 2 wherein the pain treated is neuropathic pain.

6. A method according to claim 1 2 wherein the pain treated is cancer pain.

7. A method according to claim 1 2 wherein the pain treated is postoperative pain.

8. A method according to claim 1 2 wherein the pain treated is phantom limit limb pain.

9. A method according to claim 1 2 wherein the pain treated is bum burn pain.

10. A method according to claim 1 2 wherein the pain treated is gout pain.

11. A method according to claim 1 2 wherein the pain treated is osteoarthritic pain.

12. A method according to claim 1 2 wherein the pain treated is trigeminal neuralgia pain.

13. A method according to claim 1 2 wherein the pain treated is acute herpetic and postherpetic pain.

14. A method according to claim 1 2 wherein the pain treated is causalgia pain.

15. A method according to claim 1 2 wherein the pain treated is idiopathic pain.

16. A method for treating pain comprising administering a therapeutically effective amount of (S)-3-(aminomethyl)-5-methylhexanoic acid, or a pharmaceutically acceptable salt thereof, to a human in need of said treatment.

17. A method according to claim 16 wherein the compound administered is (S)-3-(aminomethyl)-5-methylhexanoic acid.

18. A method according to claim 16 wherein the compound administered is a pharmaceutically acceptable salt of (S)-3-(aminomethyl)-5-methylhexanoic acid.

19. A method according to claim 17 wherein the pain treated is chronic pain.

20. A method according to claim 17 wherein the pain treated is selected from the group consisting of inflammatory pain, neuropathic pain, cancer pain, postoperative pain, and idiopathic pain.

21. A method according to claim 17 wherein the pain treated is neuropathic pain.

22. A method according to claim 17 wherein the pain treated is diabetic neuropathic pain.

23. A method according to claim 17 wherein the pain treated is acute herpetic pain.

24. A method according to claim 17 wherein the pain treated is postherpetic pain.

25. A method according to claim 17 wherein the pain treated is fibromyalgia pain.

Referenced Cited
U.S. Patent Documents
3471548 October 1969 Keberle et al.
4024175 May 17, 1977 Satzinger et al.
4087544 May 2, 1978 Satzinger et al.
4123438 October 31, 1978 Geurts et al.
4322440 March 30, 1982 Fish et al.
4479005 October 23, 1984 Kleschick
5023269 June 11, 1991 Robertson et al.
5025035 June 18, 1991 Wallace
5051448 September 24, 1991 Shashoua et al.
5084479 January 28, 1992 Woodruff
5104869 April 14, 1992 Albright et al.
5492927 February 20, 1996 Gitter et al.
5510381 April 23, 1996 Pande
5563175 October 8, 1996 Silverman et al.
5599973 February 4, 1997 Silverman et al.
5608090 March 4, 1997 Silverman et al.
5616793 April 1, 1997 Huckabee et al.
5629447 May 13, 1997 Huckabee et al.
5637767 June 10, 1997 Grote et al.
5684189 November 4, 1997 Silverman et al.
5710304 January 20, 1998 Silverman et al.
5792796 August 11, 1998 Woodruff et al.
5840956 November 24, 1998 Grote et al.
5847151 December 8, 1998 Silverman et al.
5929088 July 27, 1999 Horwell et al.
5998435 December 7, 1999 Horwell et al.
6020370 February 1, 2000 Horwell et al.
6028214 February 22, 2000 Silverman et al.
6046353 April 4, 2000 Grote et al.
6054482 April 25, 2000 Augart et al.
6103932 August 15, 2000 Horwell et al.
6117906 September 12, 2000 Silverman et al.
6127418 October 3, 2000 Bueno et al.
6140366 October 31, 2000 Silverman et al.
6194459 February 27, 2001 Akunne et al.
6197819 March 6, 2001 Silverman et al.
6242488 June 5, 2001 Bueno et al.
6255345 July 3, 2001 Silverman et al.
6262120 July 17, 2001 Silverman et al.
6291526 September 18, 2001 Silverman et al.
6306910 October 23, 2001 Magnus
6326374 December 4, 2001 Magnus et al.
6329429 December 11, 2001 Schrier
6342529 January 29, 2002 Silverman et al.
6359005 March 19, 2002 Pande
6359169 March 19, 2002 Silverman et al.
6372792 April 16, 2002 Chouinard
6414024 July 2, 2002 Silverman et al.
6426368 July 30, 2002 Bueno et al.
6436974 August 20, 2002 Belliotti et al.
6451857 September 17, 2002 Hurtt et al.
6521650 February 18, 2003 Belliotti et al.
6525096 February 25, 2003 Silverman et al.
6544998 April 8, 2003 Mylari
6566400 May 20, 2003 Akunne et al.
6579879 June 17, 2003 Mylari
6593368 July 15, 2003 Magnus-Miller et al.
6596900 July 22, 2003 Blakemore et al.
6605745 August 12, 2003 Hoge et al.
6642398 November 4, 2003 Belliotti et al.
6680343 January 20, 2004 Angello
6689915 February 10, 2004 Hoge et al.
6703522 March 9, 2004 Blakemore et al.
6713490 March 30, 2004 Kawamura
6720348 April 13, 2004 Mylari
6730674 May 4, 2004 Martin et al.
6750171 June 15, 2004 Hoge et al.
6849629 February 1, 2005 Mylari
6855849 February 15, 2005 Hoge et al.
6887902 May 3, 2005 Schrier
6891059 May 10, 2005 Burk et al.
6894047 May 17, 2005 Mylari
6924377 August 2, 2005 Blazecka et al.
6942876 September 13, 2005 Magnus-Miller et al.
6992109 January 31, 2006 Segal et al.
7022678 April 4, 2006 Hurley et al.
7026505 April 11, 2006 Dooley et al.
7030119 April 18, 2006 Barrett et al.
7053122 May 30, 2006 Maw et al.
7067262 June 27, 2006 Su
7071339 July 4, 2006 Belmont et al.
7074814 July 11, 2006 Para et al.
7122683 October 17, 2006 Fish et al.
7138406 November 21, 2006 Chantigny et al.
7138542 November 21, 2006 Dooley et al.
7141695 November 28, 2006 Przewosny et al.
7164034 January 16, 2007 Dooley et al.
7205295 April 17, 2007 Barvian et al.
7214824 May 8, 2007 Inoue et al.
7217721 May 15, 2007 Basford et al.
7230135 June 12, 2007 Hoge, II et al.
7235363 June 26, 2007 Bertelli et al.
7235657 June 26, 2007 Li
7256216 August 14, 2007 Kulkarni et al.
7279486 October 9, 2007 Hashizume et al.
7309719 December 18, 2007 Aomatsu
7354925 April 8, 2008 Hashizume et al.
7381747 June 3, 2008 Dooley et al.
7390931 June 24, 2008 Hoge, II
7414156 August 19, 2008 Hoge, II et al.
7419981 September 2, 2008 Field et al.
7423054 September 9, 2008 Yuen
7425569 September 16, 2008 Bradley et al.
7482375 January 27, 2009 Bradley et al.
7485636 February 3, 2009 Yuen
7491835 February 17, 2009 Donevan et al.
7507742 March 24, 2009 Rawson et al.
7514457 April 7, 2009 Inoue et al.
7547714 June 16, 2009 Cheng et al.
7553877 June 30, 2009 Chantigny et al.
7566739 July 28, 2009 Hanazawa et al.
7569572 August 4, 2009 Bell et al.
7572797 August 11, 2009 Denton et al.
7572799 August 11, 2009 Bell et al.
7572910 August 11, 2009 Mylari
7579471 August 25, 2009 Basford et al.
7589109 September 15, 2009 Uchida et al.
7595329 September 29, 2009 Ando et al.
7598231 October 6, 2009 Cheng et al.
7598393 October 6, 2009 Kon-I et al.
7612226 November 3, 2009 Graham et al.
7622589 November 24, 2009 Hanazawa et al.
7649004 January 19, 2010 Lane et al.
7659305 February 9, 2010 Rawson
7659394 February 9, 2010 Barta et al.
20010036943 November 1, 2001 Coe et al.
20020058706 May 16, 2002 Schrier et al.
20020072533 June 13, 2002 Schrier et al.
20030045449 March 6, 2003 Lowe et al.
20030045500 March 6, 2003 Magnus et al.
20040002543 January 1, 2004 Magnus et al.
20040006073 January 8, 2004 Dooley
20040092522 May 13, 2004 Field et al.
20040097405 May 20, 2004 Schrier et al.
20040132636 July 8, 2004 Dooley et al.
20040138305 July 15, 2004 Taylor, Jr. et al.
20040143014 July 22, 2004 Bertrand et al.
20050004106 January 6, 2005 Romano
20050004177 January 6, 2005 Roark
20050059654 March 17, 2005 Arneric et al.
20050059715 March 17, 2005 Dooley et al.
20050065176 March 24, 2005 Field et al.
20050148573 July 7, 2005 Katsu et al.
20050171203 August 4, 2005 Meyer-Wonnay et al.
20050182049 August 18, 2005 Howard
20050222464 October 6, 2005 Hoge
20050228190 October 13, 2005 Bao et al.
20050277672 December 15, 2005 Ando et al.
20050283023 December 22, 2005 Hu et al.
20060003344 January 5, 2006 Houseknecht et al.
20070191350 August 16, 2007 Field et al.
20070191462 August 16, 2007 Hettiarachchi et al.
20070196905 August 23, 2007 Burns et al.
20080293746 November 27, 2008 Gunn
20090036487 February 5, 2009 Field et al.
20090156677 June 18, 2009 Aomatsu
20090170897 July 2, 2009 Corradini et al.
20090318451 December 24, 2009 Ackley
20100035880 February 11, 2010 Hanazawa et al.
Foreign Patent Documents
1304080 June 1992 CA
2265615 March 1998 CA
2530904 June 2007 CA
024965 April 1983 EP
088593 September 1983 EP
181833 May 1986 EP
0 300 448 January 1989 EP
0 353 350 February 1990 EP
0368766 May 1990 EP
399949 November 1990 EP
0414263 February 1991 EP
419247 March 1991 EP
0446 570 September 1991 EP
2126224 March 1984 GB
49-40460 November 1974 JP
07 215863 August 1995 JP
WO 85/00520 February 1985 WO
WO92/09560 June 1992 WO
WO92/009560 June 1992 WO
WO 92/14443 September 1992 WO
WO 93/12811 July 1993 WO
WO93/23383 November 1993 WO
WO93/023383 November 1993 WO
WO 94/25016 November 1994 WO
WO 95/32730 December 1995 WO
WO 96/003122 February 1996 WO
WO96/011680 April 1996 WO
WO96/015782 May 1996 WO
WO96/021661 July 1996 WO
WO96/026929 September 1996 WO
WO 97/29101 August 1997 WO
WO 97/39768 October 1997 WO
WO 98/58641 December 1998 WO
WO99/059572 November 1999 WO
WO99/059573 November 1999 WO
WO00/061234 October 2000 WO
WO01/001983 January 2001 WO
WO01/024791 April 2001 WO
WO01/024792 April 2001 WO
WO2005/102389 November 2005 WO
WO2005/102390 November 2005 WO
WO06/008640 January 2006 WO
WO06/092692 September 2006 WO
WO06/123247 November 2006 WO
WO07/052125 May 2007 WO
WO07/102058 September 2007 WO
Other references
  • Lever et al (in Annual Reports in Medicinal Chemistry, vol. 19: p. 5, 1984).
  • Berge et al (J Pharm Sci 66:1-19, 1977).
  • Plea (response) of ANVISA and INPI relating to Brazillan patent application P19710536-8 dated Oct. 5, 2009 (date on last page) (in Portugese).
  • English Translation of document C1.
  • Notificacao n. * 204/09 for PI 9710536-8 (Notification Regarding the Decision to Deny Prior Approval by ANVISA on May 13, 2009) (in Portuguese).
  • English Translation of document C3.
  • Andruszkiewcz et al., Chemoenzymatic Synthesis of (R)- And (S)-4-Amino-3-Methylbutanoic Acids; Synthetic Comm. (1990) 20(1): 159-166.
  • Audus et al., Characteristics of the Large Neutral Amino Acid Transport System of Bovine Brain Microvessel Endothelial Cell Monolayers, Journal Of Neurochemistry (1986) 47(2):484-488.
  • Bartoszyk et al, Gabapentin, Current Problems In Epilepsy, New Anticonvlsant Drugs (1986) 4:147-163.
  • Burger, Alfred, A Guide to the Chemical Basis of Drug Design, John Wiley & Sons, Inc., A Wiley-Interscience publication; Chapter 2 Recent Active Research Areas; (1983) 37-91.
  • Buu et al., Biological Actions in vivo of two y-aminobutyric acid (GABA) Analogues : B-chloro Gaba and B-Phenyl Gaba ; Br. J. Pharmacol. (1974) 52 : 401-406.
  • Colonge et al, Preparation of 2-Pyrrolidones and y-Amino Acids, Bulletin De La Societe Chimique De France (1962) 598-603.
  • Crawford et al., Gabapentin as an antiepileptic drug in man, J. Neurology Neurosurgery and Psychiatry, (1987) 50:682-686.
  • Korsgaard, “Bacolfen (Lioresal) in the treatment of neuroleptic-induced tardive dyskinesia”, (1976) 54:17-24.
  • Karlsson, et al; Effect of the Convulsive agent 3-mercaptopropionic acid on the levels of GABA, other amino acids and glutamate decarboxylase in different regions of the rat brain; Biochem. Pharmacol (1974) 23:3053-3061.
  • Krall, R.L., et al., Antiepileptic drug development: II.Anticonvulsant drug screening, Epilepsia (1978) 19:409-428.
  • Litchfield, J.T. and Wilcoxon, F.; A simplified method of evaluating dose-effect experiments, Journal Of Pharmacology And Experimental Therapeutics (1949) 96: 99-113.
  • Loscher and Schmidt, Which animal models should be used in the search for new antiepileptic drugs? A proposal based on experimental and clinical considerations, Epilepsy Research (1988) 2(3): 145-181.
  • Komissarov, S.I., “Non-opiate Subarachnoidal Analgesia Induced by GABA-Positive Substances”, Farmakologiia, Toksikologiia Problemy Toksikologii (1985) 48(4): 54-58.
  • Jurna et al., Antikonvulsiva beim Nervenschmerz(Anticonvulsant agents in neuralgic pain), Abstract, Institut fur Pharmakologie und Toxikologie der Universitat des Saarlandes, W-6650, Homburg/Saar, Bundesrepublik Deutschland ; Der Schmerz (1992) 6: 146-149.
  • Kopelvich, “Advances in the Search for Medicinal Drugs Based on y-Aminobutyric Acid”, Russian Chemical Reviews (1979) 48(7): 679-691.
  • Zobacheva et al.; The Interaction of Nitroolefins With Malonic Dimethyl Ester; Higher Education Scientific Reports; Chemistry and Chemical Technology; No. 4; pp. 740-742 (1958).
  • Nicoll, “The Effect of Conformationally Restricted Amino Acid Analogues on the Frog Spinal Cord in vitro”, Br. J. Pharm., (1977) 59: 303-309.
  • Pardridge, Strategies for Drug Delivery through the Blood Brain Barrier, Directed Drug Delivery, Borchardt, Repta and Stella, eds. Humana Press, Clifton, New Jersey, (1985) pp. 83-96.
  • Perekalin & Zobacheva, “Synthesis Of y-Amino Acids And Pyrrolidones,” J. Gen. Chem. USSR (1959) 29: 2865-2869.
  • Pierdda et al., Effect of stimulus intensity on the profile of anticonvulsant activity of phenytoin, ethosuximide and valproate, Journal Of Pharmacology And Experimental Therapeutics (1985) 232(3): 741-745.
  • Purpura et al., Structure-Activity Determinants of Pharmacological Effects of Amino Acids and Related Compounds on Central Synapses, J. Neurochem. (1959) 3:238-268.
  • Saletu et al., “Evaluation of encephalotropic and psychotropic properties of gabapentin in man by pharmaco-EEG and psychometry”, Int. J. Clin. Pharmacol Ther Toxcol, (Jul. 1986) 24(7):362-373.
  • Silverman et al., Substituted 4-Aminobutanoic Acids: Substrates For y-Aminobutyric Acid α-Ketoglutaric Acid Aminotransferase, J. Bio. Chem.(Nov. 1981) 256(22): 11565-11568.
  • Silverman et al., Substrate Stereospecificity and Active Site Topography of y-Aminobutyric Acid Aminotransferase for β-Aryl-y-aminobutyric Acid Analogues, J. Bio. Chem., (1987) 262(7): 3192-3195.
  • Smith et al., Kinetics of Neutral Amino Acid Transport Across the Blood-Brain Barrier, Journal Of Neurochemistry, (1987) 49(5): 1651-1658.
  • Swinyard et al., “General Principles: Experimental Selection, Qualification, and Evaluation of Anticonvulsants,” Antiepileptic Drugs, Third Edition, edited by R. Levy et al., Raven Press, Ltd., New York, (1989) 85-102.
  • Taylor, et al., “3-Alkyl GABA and 3-Alkylglutamic Acid Analogues: Two New Classes of Anticonvulsant Agents”, Epilepsy Res., (1992) 11: 103-110.
  • Zapp, “Postpoliomyelitis Pain Treated with Gabapentin”, American Family Physician, (1996) 53(8), pp. 2442 and 2445.
  • Baxter, C.F and Roberts, E.; The y-Aminobutyric Acid-a-Ketoglutaric Acid Transaminase of Beef Brain; J. Biol. Chem. (1958) 233(5): 1135-1139.
  • Butterworth J, et al, Phosphate-Activated Glutaminase in Relation to Huntington's Disease and Agonal State ; J. Neurochem. (1983);41(2):440-447.
  • Campbell, et al., Clinical trial of carbamazepine in trigeminal neuralgia, J. Neuro. Neurosurg. Psychiat. (1966) 29: 265-267.
  • Carvajal G, et al, Anticonvulsive Action of Substances Designed as Inhibitors of y-Aminobutyric Acid-a-Ketoglutaric Acid Transaminase ; Biochem. Pharmacol. (1964) 13:1059-1069.
  • Chadwick, Recent Advances in Epilepsy, Pedley T A, Meldrum B S, (eds.) Churchill Livingstone, New York (1991) 5: 211-222.
  • Gee, et al., The Novel Anticonvulsant Drug, Gabapentin (Neurontin), Binds to the α2 σSubunit of a Calcium Channel , J Biol. Chem., (Mar. 1996) 271(10), 5768-5776.
  • Sobocinska et al.; Resolution of Racemic β-Phenyl-y-Aminobutyric Acid Into Its Enantiomers and Determination of Their Absolute Configuration; Roczniki Chemii 48; pp. 461-465 (1974).
  • Hayashi; The inhibitory action of β-Hydroxy-y-Aminobutyric Acid Upon the Seizure following Stimulation of the Motor Cortex of the Dog; J. Physiol. (London) (1959) 145:570-578.
  • A.F. Casy; Stereochemistry and Biological Activity; (7) from Medicinal Chemistry (3rd Edition) Part 1; Editor Alfred Burger; Wiley-Interscience; Cover pages and pp. 81-107 (1970).
  • Iversen, et al, Psychiatry Research (1974)11:255-256.
  • Janssens de Varebeke , et al, Effect of Milacemide, A Glycinamide Derivative, on the rat Brain y-Aminobutyric Acid System ; Biochem. Pharmacol. (1983)32(18):2751-2755.
  • Kaplan, “New Anticonvulsants: Schiff Bases of y-Aminobutyric Acid and y-Aminobutyramide”, J. Med. Chem., (1980)23: 702-704.
  • Loscher , Anticonvulsant and Biochemical Effects of Inhibitors of GABA Aminotranserase and Valproic Acid During Subchronic Treatment in Mice; Biochem. Pharmacol. (1982) 31(5):837-842.
  • Mackin, Medical and pharmacologic management of upper extremity neuropathic pain syndromes; J Hand Therapy (Apr./Jun. 1997)10(2) 96-109.
  • Mao et al., Gabapentin in Pain Management; Anesth Analg (2000)91 :680-7.
  • McGeer, et al., GABA and Glutamate Enzymes; Glutamine, Glutamate, and GABA in the Central Nervous System; Eds Liss: New York (1983) 3-17.
  • McGeer et al., The GABA System and Function of the Basal Ganglia: Huntington's Disease; GABA in Nervous System Function; Roberts et al., Eds., Raven Press: New York (1976) 487-495.
  • Meldrum, et al, Neuronal Inhibition Mediated by GABA and Patterns of Convulsions in Baboons with Photosensitive Epilepsy (Papio Papio); Epilepsy; Harris et al., Eds., Churchill Livingston (1974) 55-64.
  • Mellick et al., The use of gabapentin in the treatment of reflex sympathetic dystrophy and a phobic disorder. Am J Pain Manage (1995) 5(1):7-9.
  • Phillips et al., The effects of sodium valproate on y-aminobutyrate metabolism and behaviour in naive and ethanolamine-o-sulphate pretreated rats and mice ; Biochem. Pharmacol. (1982) 31(13):2257-2261.
  • Roberts et al., GABA in Nervous System Function, Raven Press: New York, 1976 (Table of Contents only).
  • Rock et al., Gabapentin actions on ligand- and voltage-gated responses in cultured rodent neurons. Epilepsy Res. (1993) 16: 89-98.
  • Shashoua et al., y-Aminobutyric Acid Esters. 1. Synthesis, Brain Uptake, and Pharmacological Studies of Aliphatic and Steroid Esters of y-Aminobutyric Acid; J. Med. Chem. (1984)27:659-664.
  • Silverman R. B., Mechanism-Based Enzyme Inactivation: Chemistry and Enzymology, vol. I and II, CRC: Boca Raton (1988).
  • Sist et al., Gabapentin for idiopathic trigeminal neuralgia: report of two cases. Neurology, (1997)48: 1467-1471.
  • Spokes, GABA in Huntington's Chorea, Parkinsonism and Schizophrenia; Adv. Exp. Med. Biol. (1978)123:461-473.
  • Steinman et al.; Narrative Review:The Promotion of Gabapentin: An Analysis of Internal Industry Documents; Annals of Internal Medicine, 145(4), pp. 284-293 (2006).
  • Tomson, et al., Carbamazepine in Trigeminal Neuralgia: Clinical Effects in Relation to Plasma-Concentration, Upsala J. Med. Sci., Suppl., (1980) 31: 45-46.
  • Xiao and Bennett, in Gabapentin Relieves Abnormal Pains in A Rat Model Of Painful Peripheral Neuropathy, Society For Neuroscience Abstracts (1995) 21(2):356.17.
  • Waldman S D, Tutorial 28: Evaluation and Treatment of Trigeminal Neuralgia. Pain Digest (1997) 7(1):21-24.
  • Wetzel et al., Use of gabapentin in pain management. The Annals of Pharmacotherapy; (Sep. 1997)31: 1082-3.
  • Wu et al., Abnormalities of Neurotransmitter Enzymes in Huntington's Chorea; Neurochem. Res. (1979)4(5):575-586.
  • Yurovskaya and Borschheva, in Psychoemotional Regulation and Labor Pain Relief During Phenibut Administration, Voprosy Okhrany Materinstava I Detstva (1990) 35(5):55-58.
  • Yoon Kim et al.; Glutamic Acid Analogs. The Synthesis of 3-Alkylglutamic Acids and 4-Alkylpyroglutamic Acids; J. Med. Chem., 8(4), 509-513 (1965).
  • Cronin et al.; Gas Chromatographic-Mass Spectral Analysis of the Five-Carbon β-, y-, and σ-Amino Alklanoic Acids, Analytical Biochemistry 124, pp. 139-149 (1982).
  • Silverman, “4-Amino-2(substituted methyl)-2-butenoic Acids: Substrates and Potent Inhibitors of GABA Aminotransferase”, J. Med. Chem., (1986)29: 764-770.
  • Silverman, “From Basic Science to Blockbuster Drug: The Discovery of Lyrica”, Angew. Chem. Int. Ed., (2008) 47:3500-3504.
  • Andruszkiewicz, et al., “A Convenient Synthesis of 3-Alkyl-4-aminobutanoic Acids”, Synthesis, Journal of Synthetic Organic Chemistry (Dec. 1989) 953-955.
  • Field, et al., “Gabapentin(Neurontin) and S-(+)-3-Isobutylgaba Represent a Novel Class of Selective Antihyperalgesic Agents”, British J. Pharmacol., (1997) 121: 1513-1522.
  • McQuay, et al., “Anticonvulsant Drugs for the Management of Pain: A Systematic Review”, BMJ, (Oct. 1995) 311: 1047-1052.
  • Mellick, et al., “Gabapentin in the Management of Reflex Sympathetic Dystrophy”; J. Pain Symptom Management, (1995) 10 (4): 265-266.
  • Mellick, “Successful Treatment of Reflex Sympathetic Dystrophy with Gabapentin”; Am. J. Emerg. Med., (1995) 96.
  • Mellick, et al., “Reflex Sympathetic Dystrophy Treated with Gabapentin”, Arch. Phys. Med. Rehabil., (1997) 78: 98-105.
  • Rosner, et al., “Gabapentin Adjunctive Therapy in Neuropathic Pain States”, Clin. J. Pain, (1996) 12 (1): 56-58.
  • Segal, et al., “Gabapentin as a Novel Treatment for Postherpetic Neuralgia”, American Academy of Neurology, (1996) 46(4): 1175-1176.
  • Suman-Chauhan, et al., “Characterisation of [3H]gabapentin Binding to a Novel Site in Rat Brain; Homogenate Binding Studies”, Eur. Jr. Pharmacol., (1993) 244 (3): 293-301.
  • Taylor, et al., “Potent and Stereospecific Anticonvulsant Activity of 3-Isobutyl GABA Relates to in Vitro Binding at a Novel Site Labeled by Tritiated Gabapentin”, Epilepsy Res., (1993)14: 11-15.
  • Thurlow, et al., “[3H]Gabapentin May Label a System-L-Like Neurtral Amino Acid Carrier in Brain”, European Journal of Pharmacology, (1993)247:341-345.
  • Certified English Language Translation of document B1.
  • Certified English Language Translation of document C6.
  • Certified English Language Translation of document C13.
  • Certified English Language Translation of document C14.
  • Certified English Language Translation of document C16.
  • Certified English Language Translation od document C63.
  • U.S. Appl. No. 60/559,194 .
  • U.S. Appl. No. 10/018,616 , Brummel et al.
  • U.S. Appl. No. 60/142,215 .
  • U.S. Appl. No. 10/089,958 , Hughes et al.
  • U.S. Appl. No. 60/158,271.
  • U.S. Appl. No. 10/089,819, Hughes et al.
  • U.S. Appl. No. 08/445,398, Woodruff et al.
  • U.S. Appl. No. 08/25,5143.
  • U.S. Appl. No. 08/924,779.
  • Goodman and Gilman's The Pharmacological Basis of Therapeutics, 8th Edition, Chapter 19, Figure 19-1 (1990).
  • Presentation at the Indiana Chapter RSDS [Reflex Sympathetic Dystrophy Syndrome] Symposium on Aug. 13, 1994.
  • Poster presentation entitled “Successful Treatment of Reflex Sympathetic Dystrophy with Gabapentin (Neurontin)”, at the 13th Annual Scientific Meeting of the American Pain Society in Miami Beach, Florida, on Nov. 10-13, 1994.
  • Sist et al., Gabapentin for Idiopathic Trigeminal Neuralgia; Report of Two Cases, Neurology, May 1997 48:1467.
  • Burger A., Medicinal Chemistry, John Wiley & Sons, 1970, pp. 81, 83.
  • Handley, S.L. and Singh, L. (1985). Modulation of 5-hydroxytryptamine-induced head-twitch response by drugs acting at GABA and related receptors. Br. J. Pharmacol., 86, 297-303.
  • Singh, L. Heaton, C.J.P., Rea, P.J. and Handley, S.L. (1986), Involvement of noradrenaline in potentiation of the head-twitch response by GABA-related drugs. Psychopharmacol.88, 315-319.
  • Handley, S.L., and Singh, L. (1986) The modulation of head-twitch behaviour by drugs acting on the beta-adrenoceptors. Evidence for involvement of both beta1--and beta2-adrenoceptors. Psychopharmacol.88, 320-324.
  • Handley, S.L. and Singh, L. (1986). Involvement of Locus Coeruleus in the potention of the quipazine head-twitch response by diazepam and beta-adrenceptor agonists. Neuropharmacol., 25, 1315-1321.
  • Handley, S.L. and Singh, L. (1986). Chronic antidepressant treatment reduces central β-adrenoceptor sensitivity in a behaviour test. Eur. J. Pharmacol. 127, 97-103.
  • Singh, L. and Handley, S.L. (1987). Behavioural evidence for an interdependence between GABAa receptors and Beta2-adrenoceptors. Eur. J. Pharmacol., 135, 419-421.
  • Handley, S.L., and Singh, L. (1986) Neurotransmitters and shaking behaviour-more than a ‘gut-bath’ for the brain? Trends Pharmacol. Sci., 7, 324-328.
  • Tricklebank, M.D., Singh, L., Oles, R.J., Wong, E.H.F. and Iversen, S.D. (1987). A role for receptors of N-methyl-D-aspartic acid in the discriminative stimulus properties of phencyclidine. Eur. J. Pharmacol., 141, 497-501.
  • Price, B.W., Ahier, R.G., Middlemiss, D.N., Singh, L., Tricklebank, M.D., and Wong, E.H.F. (1988). In vivo labeling of the NMDA receptor channel complex by [3H]MK-801. Eur. J. Pharmacol., 158, 279-282.
  • Iversen, S.D., Singh, L., Oles, R.J., Preston, C. and Tricklebank, M.D. (1988). Psychophamacological profile of the N-methyl-D-aspartate (NMDA) receptor antagonist, MK-801. In Sigma and Phencyclidine-Like Compounds as Molecular Probes in Biology, edited by E.F. Domino and J.M. Kamenka pp. 373-382.
  • Tricklebank, M.D., Singh, L., Oles, R.J., Preston, C. and Iversen, S.D. (1989). The behavioural effects of MK-801: a comparison with antagonists acting non-competitively and competively at the NMDA receptor. Eur. J. Pharmacol., 167, 127-135.
  • Singh, L., E.H.F., Kesingland, A. and Tricklebank, M.D. (1990). Evidence against an involvement of the haloperidol-sensitive sigma recognition site in the discriminative stimulus properties of (+)-N-allyl-normeta30cine (+)-SKF-10,047). Br. J. Pharmacol.99, 145-151.
  • Singh, L., Oles, R.J. and Tricklebank, M.D. (1990). Modulation of seizure susceptibility in the mouse by the strychnine-insensitive glycine recognition site of the NMDA receptor/ion channel complex. Br. J. Pharmacol., 99, 285-288.
  • Singh, L. Donald, A.E., Foster, A.C., Hutson, P.H., Iversen, L.L., Iversen, S.D., Kemp, J.A., Leeson, P.D., Marshall, G.R., Oles, R.J., Priestley, T., Thorn, L., Tricklebank, M.D., Vass, C.A. and Williams, B.J. (1990), Enantiomers of HA-966 (3-amino-1-hydroxpyrrolid-2-one) exhibit distinct central nervous system effects: (+) —HA-966 is a selective glycine/N-methyl-D-aspartate receptor antagonist, but (−)-HA 966 is a potent gamma-butyrolactone-like sedative, Proc. Natl. Acad. Sci., U.S.A. 87, 347-351.
  • Singh, L., Menzies, R. and Tricklebank, M.D. (1990). The discriminative stimulus properties of (+)-HA-966, an antagonist at the glycine/N-methyl-D-aspartate receptor. Eur. J. Pharmacol., 186, 129-132.
  • Tricklebank, M.D., Honore, T., Iversen, S.D., Kemp, J.A., Knight, A.R., Marshall, G.R., Rupniak, N.M.J., Singh, L., Tye, S., Watjen, F. and Wong, E.H.F. (1990). The pharmacological properties of the imidazobenzodiazepine, FG 8205, a novel partial agonist at the benzodiazepine receptor. Br. J. Pharmacol., 101, 753-761.
  • Thompson, W. J., Anderson, P.S., Britcher, S.F., Lyle, T.A., Thies, J.E. Magill, C.A., Varga, S.L., Schwering, J.E, Lyle, P.A., Christy, M.E., Evans, B.E., Colton, C.D., Holloway, M.K., Springer, J.P., Hirshfield, J.M., Ball, R.G., Amato, J.S., Larsen, R.D., Wong, E.H.F., Kemp, J.A., tricklebank, M.D., Singh, L., Oles, R.J., Priestly, T., Marshall, G.R, Knight, A.R., Middlemiss, D.N., Woodruff, G.N., and Iversen, L.L. (1990). Synthesis and pharmacological evaluation of a series of dibenzo([a,s])cycloalkenimines as N-methyl-D-aspartate antagonists. J. Med. Chem., 33, 789-808.
  • Singh, L. Oles, R. and Woodruff, G. (1990). In vivo interaction of a polyamine with the NMDA receptor. Eur. J. Pharmacol., 180, 391-392.
  • Singh, L., C.A., Hunter, J.C., Woodruff, G.N. and Hughes, J. (1990). The anticonvulsant action of CI-977, a selective kappa-opioid receptor agonist: a possible involvement of the glycine/NMDA receptor complex. Eur. J. Pharmacol., 191, 477-480.
  • Singh, L., Lewis A.S., Field, M.J., Hughes, J. and Woodruff, G.N. (1991). Evidence for an involvement of the brain cholecystokinin B receptor in anxiety. Proc. Natl. Acad. Sci., U.S.A. 88, 1130-1133.
  • Singh, L., Oles, R.J., Vass, C.A. and Woodruff, G.N. (1991). A slow intravenous infusion of N-methyl-DL-aspartate as a seizure model in the mouse. J. Neurosci. Meth., 37, 227-232.
  • Hill, D.R., Singh, L., Boden, P., Pinnock, R., Woodruff, G.N. and Hughes, J. (1992). Detection of CCK receptor subtypes in mammalian brain using selective non-peptide antagonists. In Multiple Cholecystokinin Receptors in the CNS. Edited by Dourish, C. T., Cooper, S.J., Iversen, S.D. and Iversen L.L. Oxford University Press, pp. 57-76.
  • Woodruff, G.N., Hill, D., Boden,P. , Pinnock, R., Singh, L. and Hughes, J. (1991). Functional role of brain CCK receptors. Neuropeptides, 19, (Suppl.), 45-56.
  • Singh., L., Field, M.J., Hughes, J., Oles, R.J., Vass, C.A. and Woodruff, G.N. (1991). The behavioural properties of CI-988, a selective CCK-B receptor antagonist. Br. J. Pharmacol., 104, 239-245.
  • Tricklebank, M.D., Singh, L., Jackson, A. and Oles, R.J. (1991). Evidence that a proconvulsant action of lithium is mediated by inhibition of myo-inositol phosphatase in mouse brain. Brain Res., 558, 145-148.
  • Singh, L., Field, M.J., Vass, C.A., Hughes, J. and Woodruff, G.N. (1992). The antagonism of benzodiazepine withdrawal effects by the selective cholecystokinin-B receptor antagonist CI-988. Br. J. Pharmacol., 105, 8-10.
  • Boden, P.R., Higgenbottom, M., Hill, D.R., Horwell, D.C., Hughes, J., Rees, D.C., Roberts, E., Singh, L., Suman-Chauhan, N., and Woodruff, G.N. (1993). Cholecystokinin dipeptoid antagonists: Design, synthesis and anxiolytic profile of some novel CCK-A and CCK-B selective and “mixed” CCK-A/CCK-B antagonists. J. Med. Chem., 36, 552-565.
  • Moore, K.W., Leeson, P.D., Carling, R.W., Tricklebank, M.D. and Singh, L. (1993). Anticonvulsant activity of glycine-site NMDA antagonists. 1.2-Carboxyl prodrugs of 5,7-dichlorokynurenic acid. Bioorganic& Med. Chem. Lett., 3, 61-64.
  • Hunter, J.C. and Singh, L. (1994). Role of excitatory amino acid receptors in the mediation of the nociceptive response to formalin in the rat. Neurosci. Lett.174, 217-221.
  • Hunter, J.C., Atwal, P., Woodruff, G.N. and Singh, L. (1994). Differential modulation of κ and μ opoioid antinociception by the glycine/NMDA receptor agonist D-serine. Br. J. Pharmacol., 112, 1002-1003.
  • Tricklebank, M.D., Bristow, L.J., Hutson, P.H., Leeson, P.D., Rowley, M.Saywell, K., Singh, L., Tattersall, F.D., Thorn, L. and Williams, B.J. (1994). The anticonvulsant and behavioural profile of L-687,414, a partial agonist acting at the glycine modulatory site on the N-methyl-D-aspartate (NMDA) receptor complex. Br. J. Pharmacol., 113, 729-736.
  • Singh, L., Field, M.J., Hill, D.R, Horwell, D.C., McKnight, A.T., Roberts, E., Tang, K.W. and Woodruff, G.N. (1995). Peptoid CCK receptor antagonists: pharmacological evaluation of CCKA1 CCKB and mixed CCKA/B receptor antagonists. Eur. J. Pharmacol., 286, 185-191.
  • Singh, L., Oles, R.J., Field, M.J., Atwal, P. Woodruff, G.N. and Hunter, J.C., (1996). Effect of CCK receptor antagonists on the antinociceptive, reinforcing and gut motility properties of morphine. Br. J. Pharmacol., 118, 1317-1325.
  • Singh, L., Field, M.J., Hunter, J.C., Oles, R.J. and Woodruff, G.N. (1996). Modulation of the in vivo actions of morphine by the mixed CCKA/B receptor antagonist PD 142898. Eur. J. Pharmacol.307, 283-289.
  • Singh, L., Field, M.F., Ferris, P., Hunter, J.C., Oles, R.J., Williams, R.G. and Woodruff, G.N. (1996). The Antiepileptic Agent Gabapentin (Neurontin) Possesses Anxiolytic-Like and Antinociceptive Actions that are Reversed by D-serine. Psychopharmacol., 127, 1-9.
  • Brown, J.P., Boden, O., Singh, L. and Gee, N.S. (1996). Mechanisms of action of gabapentin (Neurontin). Rev. Contem. Pharmacother., vol. 7, No. 5, 203-214.
  • Singh, L., Field, M.J., Hughes, J., Kuo, B.-S., Suman-Chauhan, N., Tuladhar, B.R., Wright, D.S. and Naylor, R.J. The tachykinin NK1 receptor antagonist PF 154075 blocks cisplatin-induced delayed emesis in the ferret. Eur. J. Pharmacol., 321, 209-216 (1997).
  • Field, M.J., Oles, R.J., Lewis, A.S., McCleary, S., Hughes, J. and Singh, L. (1997). Gabapentin (Neurontin) and S-(+)-3-isobutylgaba represent a novel class of selective antihyperalgesic agents. Br. J. Pharmacol., (1997) 121, 1513-1522.
  • Stanfa, L.C., Singh, L., Williams, R.G. and A.H. Dickenson (1997). Gabapentin (Neurontin), ineffective in normal rats, markedly reduces C-fibre evoked responses after inflammation. Neuroreport, 8, 587-590 (1997).
  • Field, M.J., Holloman, E.F., McCleary, S., Hughes, J., Singh, L. Evaluation of gabapentin and S-(+)-3-isobutylgaba in a rat model of postoperative pain, Journal of Pharmacology and Experimental therapeutics, 282, 1242-1246 (1997).
  • Handley, S.L. and Singh, L. (1984). The effect of beta adrenoceptor agonists and antagonists on head-twitch in male mice. Br. J. Pharmacol., 81, 127P.
  • Handley, S.L., and Singh, L. (1984). GABA modulates the head-twitch induced by L-5-HTP. Br. J. Pharmacol., 82, 340P.
  • Handley, S.L., and Singh, L. (1986). GABAa agonists potentiate and baclofen antogonises the L-5-HTP head-twitch. Br. J. Pharmacol., 84, 86P.
  • Handley, S.L., Mithani, S. and Singh, L. (1985). Locus Coeruleus lesions do not affect diazepam- or alpha-adrenergic modulation of operant conflict. Br. J. Pharmacol., 84, 87P.
  • Handley, S.L., Singh, A. and Singh, L. (1986). Ritanserin reduces morphine and clonidine withdrawl ticks. Br. J. Pharmacol., 89, 647P.
  • Tricklebank, M.D., Pharmacology, 1987, 299-302.
  • Iversen, S.D., Oles, R.J., Singh, L. and Tricklebank, M.D. (1986). Involvement of the haloperidol-sensitive sigma recognition site in the behaviours induced in the rat by (+)SKF-10,047. Br. J. Pharmacol., 91, 340P.
  • Tricklebank, M.D., Wong, E., Kemp, J., Singh, L., Rupniak, N., Woodruff, G.N., Iversen, S.D., Iversen, L.L. and Watjen, F. (1988). The Pharmacological profile of FG 8205, a partial agonist at the benzodiazepine receptor. Psychopharmacol., 96, 28.33.07.
  • Singh, L., Barth, T., Rupniak, N., Tricklebank, M.D. and Iversen, S.D. (1988). The tolerance and dependance potential of FG 8205, a partial agonist at the benzodiazepine receptor. Psychopharmacol., 96, 28.33.08.
  • Iversen, S.D., Singh, L., Oles, R.J. and Tricklebank, M.D. (1988). The behavioural effects of excitatory amino acid (EAA) antagonists. Psychopharmacol., 96, 530/112.
  • Tricklebank, M.D., Oles, R.J. and Singh, L. (1990). Reversal by inositol of the proconvulsant action of lithium in pilocarpine-treated mice. Br. J. Pharmacol., 99, 73P.
  • Oles, R.J., Singh, L. and Tricklebank, M.D. (1990). Differential effects on the behavioural and anticonvulsant properties of MK-801 following repeated administration in the mouse. Br. J. Pharmacol., 99, 286P.
  • Singh, L., Oles, R. and Woodruff, G.N. (1990). In vivo interaction of polyamine with the NMDA receptor. 20th Annual meeting Soc. for Neurosciences. St. Louis, U.S.A., 200.4.
  • Oles, R., Singh, L., Hughes, J. and Woodruff, G.N. (1990). The anticonvulsant action of gabapentin involves the glycine/NMDA receptor. 20th Annual meeting Soc. for Neurosciences. St. Louis, U.S.A., 221.6.
  • Singh, L., Field, M.J., Hughes, J., Vass, C.A. and Woodruff, G.N. (1991). Central administration of a CCK-B receptor agonist induces anxiety. Br. J. Pharmacol., 102, 45P.
  • Field, M.J., Hughes, J., Lewis., A.S., Oles, R.J., Singh, L., Vass, C.A and Woodruff, G.N. (1991). The anxiolytic-like actions of the selective CCK-B receptor antagonist CI-988. Br. J. Pharmacol., 102, 256P.
  • Field, M.J., Lewis, A.S., Lloyd, S. and Singh, L. (1991). Automation of the rat elevated X-maze test of anxiety. Br. J. Pharmacol., 102, 304P.
  • Saywell, K., Singh, L., Oles, R.J., Vass, C., Leeson, P.D., Williams, B.J. and Tricklebank, M.D. (1991). The anticonvulsant properties in the mouse of the glycine/NMDA receptor antagonist, L-687,414. Br. J. Pharmacol., 102, 66P.
  • Singh, L., Oles, R.J. and Woodruff, G.N. (1991). The lack of sedative properties of CI-988, a selective CCKB receptor antagonist. 21st Annual Meeting Soc. For Neurosciences. New Orleans, U.S.A.
  • Singh, L., Hughes, J., Field, M. and Woodruff, G.N.,(1992). The effects of the CCKB receptor-antagonist CI-988, on withdrawal from chronic alchol treatment. CPDD Meeting in Colorado, Jun. 20-25, 1992.
  • Singh, L., Field, M. and Woodruff, G.N. (1992). Selective CCKB but not CCKA receptor antagonists show anxiolytic-like action in the rat. British Association for Psychopharmacology Meeting in Cambridge, U.K., Aug. 2-7, 1992.
  • Field, M.J., Day, H., Vass, C.A and Woodruff, G.N. (1992). Antagonism of alchol withdrawl effects by the selective CCKB receptor antagonist CI-988. British Association for Psychopharmacology Meeting in Cambridge, U.K., Aug. 2-7, 1992.
  • Hargreaves K, Dubner R, Brown F, Flores C. Joris J: A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 32: 77-88, 1988.
  • Woolf, CJ: Evidence for a central component of post-injury pain hypersensitivity. Nature 306: 686-688, 1983.
  • Treede RD, Davis KD, Campbell JN, Raja SN: The plasticity of cutaneous hyperalgesia during sympathetic ganglion blockade in patients with neuropathic pain. Brain 115 (Pt. 2): 607-621, 1992.
  • Woolf CJ: The pathophsiology of peripheral neuropathic pain—Abnormal peripheral input and abnormal central processing. Acta Neurochirurgica 58: 125-130, 1993.
  • Qian J. Brown SD, Carlton SM: Systemic ketamine attenuates nociceptive behaviors in a rat model of peripheral neuropathy. Brain Res 715: 51-62, 1996.
  • Coderre TJ, Melzack R: The role of NMDA receptor-operated calcium channels in persistent nociception after formalin-induced tissue injury. J Neurosci. 12:3671-3675, 1992.
  • Coderre TJ Melzack R: The contribution of excitory amino acids to central sensitization and peristent nociception after formalin-induced tissue injury. J Neurosci. 12: 3665-3670, 1992.
  • Coderre TJ, Yashpal K: Intracellular messengers contributing to persistent nociception and hyperalgesia induced by L-glutamate and substance P in the rat formalin pain model. The European Journal of Neuroscience 6: 1328-1334, 1994.
  • Xu XJ, Elfvin A, Wiesenfeld-Hallin Z: Subcutaneous carrageenan, but not formalin, increases the excitability of the nociceptive flexor reflex in the rat. Neuroscience Letters 196: 116-118, 1995.
  • McMahon SB: NGF as a mediator of inflammatory pain. Philosophical Transactions Royal Society of London 351: 431-440, 1996.
  • Woolf CJ: A new strategy for the treatment of inflammatory pain. Prevention or elimination of central sensitization. Drugs 47 Suppl 5: 1-9; discussion 46-47, 1994.
  • Woolf CJ, Chong MS: Preemptive analgesia—treating postoperative pain by preventing the establishment of central sensitization. Anesthesia and Analgesia 77: 362-379, 1993.
  • Sorensen J, Bengtsson A, Backman E, Henriksson KG, Bengtsson M: Pain analysis in patients with fibromyalgia. Effects of intravenous morphine, lidocaine, and ketamine. Scandinavian Journal of Rheumatology 24: 360-365, 1995.
  • Cavanaugh JM: Neural mechanisms of lumbar pain. Spine 20: 1804-1809, 1995.
  • Seibert K, Zhang Y, Leahy K, Hauser S, Masferrer J, Perkins W, Lee L, Isakson P: Pharmacological and biochemical demonstration of the role of cyclooxygenase 2 in inflammation and pain. Proceedings of the National Academy of Sciences USA 91: 12013-12017, 1994.
  • Sosnowski M., Pain Management: physiopathology, future research and endpoints. Support Care in Cancer 1:79-88, 1993.
  • Dubuisson D., Dennis, SG. The formalin test: A quantitative study of the analgesic effects of morphine, meperidine, and brain stem stimulation in rats and cats. Pain 1977; 4: 161-74.
  • Wheeler-Aceto H., Cowan A. Standarization of the rat paw formalin test for the evaluation of analgesics. Psychopharmacology 1991; 104: 35-44.
  • Kayser V., Guilbaud G. Local and remote modifications of nociceptive sensitivity during carrageenan-induced inflammation in the rat. Pain 1987; 28: 99-107.
  • Randall L., Selitto J. A method for measurement of analgesic activity on inflamed tissue. Arch Int. Pharmacodyn, 1957; 111: 409-19.
  • Brennan TJ, Vandermeulen EP, Gebhart GF. Characterization of a rat model of incisional pain. Pain 1996; 64: 493-501.
  • Levine JD, Fields HL, Basbaum AI. Peptides and the primary afferent nociceptor. J. Neurosci. 1993; 2273-86.
  • Ueda M., Kuraishi Y., Sugimoto K., Satoh M. Evidence that glutamate is released from capsaicin-sensitive primary afferent fibers in rats: Study with on-line continuous monitoring of glutamate. Neurosci. Res. 1994; 20: 231-7.
  • Yaksh TL, Rudy TA. Chronic catheterization of the spinal subarachnoid space. Physiol. & Behav. 1976; 17: 1031-6.
  • Ochoa JL, Yarnitsky D. Mechanical hyperalgesias in neuropathic pain patients; dynamic and static subtypes. Ann. Neurol 1993; 465-72.
  • Calcutt NA, Jorge MC, Yaksh TL, Chaplan SR. Tactile allodynia and formalin hyperalgesia in streptozocin-diabetic rats: Effects of insulin, aldose reductase inhibition and lidocaine. Pain 1996; 68: 293-9.
  • Bennett GJ, Xie Y-K. A peripheral monooneuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988; 33: 87-107.
  • Kim SH, Chung JM. An experimental model of peripheral neuropathy produced by segmented spinal nerve ligation. Pain 1992; 50: 355-63.
  • Wooley, PH. Collagen-induced arthritis in the mouse. Methods in Enzymology 1988; 162: 361-373.
  • Holmdahl R., Tarkowski A., Jonsson R. Involvement of macrophages and dendritic cells in synovial inflammation of collagen induced arthritis in DBA/1 mice and spontaneous arthritis in MRL/Lpr mice. Autoimmunity 1991; 8: 271-280.
  • Hill DR, Suman-Chauhan N., Woodruff GN. Localization of [3H]-gabapentin to a novel site in rat brain: Autoradiographic studies. Eur. J. Pharmacol. 1993; 244: 303-9.
  • Kocsis JD, Honmou O. Gabapentin increases GABA-induced depolarization in rat neonatal optic nerve. Neuroscience Letters 1994; 169: 181-4.
  • Ragsdale DS, Scheuer T., Catterall WA. Frequency and voltage-dependent inhibition of type IIA Na+ expressed in a mammalian cell line, by local anesthetic, antiarrhythmic, and anticonvulsant drugs. Molec. Pharmacol. 1991; 40: 756-65.
  • Moertel, et al., “Relief of Pain by Oral Medications,” Analgesic Combinations, vol. 229, No. 1, Jul. 1, 1974.
  • Sawynok, Jana, “GABAergic Mechanisms of Analgesia: An Update,” Pharmacology Biochemistry and Behavior, vol. 26, pp. 463-474 (1987).
  • Visceral Pain: a review of experimental studies, Pain 41: 167-234, 1990.
  • Mayer & Raybould, Role of Visceral Afferent Mechanisms in Functional Bowel Disorders, Gastroenterology 1990 89:1688-1704.
  • Mayer & Gebhart, Basic and Clinical Aspects of Visceral Hyperalgesia, Gastroenterology 1994: 107: 271-293.
  • Miampamba, et al., “Inflammation of the colonic wall induced by formalin as a model of acute visceral pain,” Pain 57 (1994) 327-334.
  • Morteau, et al., “Experimental Colitis Alters Visceromotol Response to Colorectal Distension in Awake Rats, ” Digestive Diseases and Sciences, vol. 39, No. 6 (Jun. 1994); pp. 1239-1248.
  • Lever et al., Annual Reports in Medicinal Chemistry (vol. 19), p. 5, 1984.
  • Berge et al., J. Pharm. Sci., 66:1-9, 1977.
  • Taylor, Mechanisms of analgesia by gabapentin and pregabalin—Calcium channel α2-σ [Cavα2-σ]ligands, Pain 142 (2009) 13-16.
  • Xiao et al., Analgesia 2:267-273, 1996 (with cover page).
  • Prescribing Information for Lyrica®.
  • Allinger et al., Organic Chemistry, Chapter 6, Worth Publishers. Inc., 1971.
  • March, J. Advanced Organic chemistry; Reactions, Mechanisms and Structure, McGraw-Hill Book Company, 1968.
  • Morrison, Asymmetric Synthesis, vol. 1.1. Academic Press, 1983, Chapter 6 (Pirkle and Finn).
  • Witczuk et al., (1978)3-(p-tolyl)-4-aminobutanoic acid, synthesis, resolution into enantiomers and pharmacological activity. Pol. J. Pharmacol. Pharmacological activity. Pol. J. Pharmacol. Pharm. 30:95-103 (“Witczuk 1978”).
  • Witczuk B et al. (1980) 3-(p-chlorophenyl)-4-aminobutanoic acid—resolution into enantiomers and pharmacological activity. Pol. J. Pharmacol. Pharm. 32:187-196 (“Witczuk 1980”).
  • Eliel, Ernest L., Sterchemistry of Chemical Compounds, International Student Edition., (McGraw Book Company Inc., 1962).
  • March, J., Advanced Organic Chemistry, 3rd Edition, John Wiley & Sons, 1985; Enantiomers, Racemates and Resolutions (J. Jacques et al. eds.) pp. 378-379 (1981).
  • Norman, RO.C, Principles of Organic Synthesis, (Methuen Co., Ltd., 1968).
  • Jacques, J. Collet, A., and Wilen, S.H. Enantiomers, Racemates and Resolutions, Wiley, New York 1981.
  • ten Hoeve, W. and Wynberg, The Design of Resolving Agents: Chiral Cyclic Phosphoric Acids. H.J. Org. Chem. 1985, 50, 4508.
  • S.H. Wilen, Tables of Resolving Agents and Optical resolutions, University of Notre Dame Press, 1972.
  • Martens J. and Bhushan R. T.I.C. enantiomeric seperation of amino acids. Int. J. Pept. Protein Res. Dec. 1989; 34(6)433-44, (“Martens 1989”).
  • Maurs M. et al., (1988) Resolution of alpha-substituted amino acid enantiomers by high-performance liquid chromatography after derivitization with a chiral adduct of o-pthalaldehyde. Application to glutamic acid analogues. J. Chromatogr. 25; 440:209-215 (“Maurs 1988”).
  • Hare PE. (1988) Chiral Mobile Phases for the Enantiomeric Resolution of Amino Acids. Chromatographic Chiral Separations. (eds. Zief M. and Crane LJ.).
  • Wainer, Trends in Analytical Chemistry, 6, 125-134 (1987).
  • Railton, J. Chromatography, 402,371-373 (1987).
  • Johns, American Laboratory, 72-76 (Jan. 1987).
  • AM. Krstulovic, J. Pharm. & Biomed. Analysis, Sep. 1987.
  • AM. Krstulovic, J. Pharm. & Biomed. Analysis, 6(6-8), 1988, 641-656.
  • D. W. Armstrong, Anal Chem. 59(2), 1987, 84A-91A.
  • Hermansson, J. Chromatogr, 269, 1983, 71-80.
  • Chiralpak Chiralcel HPLC columns advertisement. J. Chromatogr., 450, No. 2., 1988; 205c.
  • Hermansson, J., Resolution of Racemic Aminoalcohols({3-Blockers), Amines and Acids as Enantiomeric Derivatives Using a Chiral a1-Acid Glycoprotein Column: , J. Chromatogr., 325, 379-384 (1985).
  • Hermansson, J., et al. Direct Liquid Chromatographic Resolution of Acidic Drugs Using a Chiral a1-Acid-Glycoprotein Column(Enantiopac), J. Liq. Chromatogr., 9 (2&3), (1986) 621-639.
  • Wainer Barkan and Schill, LC-GC 4(5), (1986) 422-430.
  • Schill, Wainer and Barkan, J. Chromatogr., 365 (1986) 73-88.
  • Debowksi, Sybilska, and Jurczak, J. Chromatogr. 237 (1982) 303-306; Bopp and Kennedy, LC-GC 6(6), 1988 514-522.
  • Bopp and Kennedy, LC-GC 6(6), (1988) 514-522.
  • Beesley, American Laboratory (May, 1985), 78-87.
  • Okamoto, Kawashima, and Hatada, J. Chromatogr. 363 (1986) 173-186.
  • Armstrong and DeMond, J. Chromatogr. Sci. 22 (1984) 411-415.
  • Schill, Wainer and Barkan, J. Liq. Chromatogr., 9(2&3), (1986) 641-666.
  • J.T. Baker Advertisements for chiral columns, 1986 and 1988.
  • Excerpts from J.T. Baker Catalog 870C; Chiralcel OD advertisement, C&EN, Jun. 20, 1988.
  • J.T. Baker advertisement, LC.GC 4(10), Oct. 1986.
  • Regis advertisement, LC.GC 4(4) Apr. 1986,
  • Prochrom preparative HPLC advertisements LC.GC, 5(5) Aug. 1987 and LC.GC, 6(2) Feb, 1988.
  • Sepragen preparative HPLC advertisements LC.GC 4(6) Jun. 1986.
  • Allan RD, et al. (1990) A New Synthesis, Resolution and in vitro Activities of(R)- and (S)-f3-phenylGABA. Tetrahedron 46(7): 2511-24 Allan 1990).
  • Olpe H, et al., Eur. J. Pharmacol 1978, 52, 133-136.
  • Belokon YN et al., (1986) Synthesis of Enantio- and Diastero-isomerically Pure β- and y-Substituted Glutamic Acids via Glycine Condensation with Activated Olefins. J. Chem. Soc. Perkin Trans. 1:1865-1872.
  • Takano S. et al., (1987) Fractional Synthesis of (R)-y-amino- β-hydroxybutanoic acid (GABOB) from (R)-epichlorohydrin, Tetrahedron Letters 28(16): 1783-1784.
  • Thaisrivongs S. et al., Renin Inhibitors, J. Med. Chem. 1987, 30, 976-982.
  • Fadel A. and Salaun J (1988) Optically Active α-alkylsuccinates from the stereoselective alkylation of chiral imide enolates. Tetrahedron Letters 29(48): 6257-6260.
  • Fromm GH and Terrence CF (1987) Comparison of L-baclofen and racemic baclofen in trigeminal neuralgia. Neurology 37: 1725-1728 (“Fromm 1987”).
  • Hill DR et al. (1993) Localization of [3H ]gabapentin to a novel site in rat brain: autoradiographic studies. European Journal of Pharmacology 244(3):303-309.
  • Allan RD et al. (1990) A New Synthesis, Resolution and in vitro Activities of(R)and (S)-(β-phenyl-GABA. Tetrahedron 46(7):2511-2524 (“Allan 1990”).
  • Weiner RS (2002) Pain Management: A Practical Guide for Clinicians (6th ed.) (“Weiner 2002”).
  • Xiao WH and Bennett GJ (1995) Synthetic w-Conopeptides Applied to the Site of Nerve Injury Suppress Neuropathic Pains in Rats, Journal of Pharmacology and Experimental Therapeutics 274(2):666-672.
  • Lapin IP et al. (1986) Antagonism of Seizures Induced by the Administration of the Endogenous Convulsant Quinolinic Acid into RatBrain Ventricles, J. Neural Transmission 65:177-185.
  • Benedito MA and Leite JR (1981) Baclofen as an Anticonvulsant in Experimental Models of Convulsions, Experimental Neurology 72:346-351.
  • Sawynok J. (1984) gabaergic Mechanisms in Antinociception, Prog. Neuro-Psychopharmacol.& Biol. Psychiat. 8:581-586 at p. 583-584.
  • Vaught JL et al. (1985) A Comparison of the Antinociceptive Responses to the GABA-Receptor Agonists THIP and Baclofen, Neuropharmacology 24(3):211-216.
  • Zarrindast MR and Djavadan M (1988) GABAA-Antagonists and Baclofen Analgesia, Gen Pharmac. 19(5):703-706.
  • Bucklett WR (1980) Irreversible Inhibitors of GABA Transaminase Induce Antinociceptive Effects and Potentiate Morphine, Neuropharmacology 19:715-722.
  • Sawynok J and Dickson C (1983) Involvement of GABA in the Antinociceptive Effect of y-acetylic GABA(GAG), an Inhibitor of GABA-Transaminase, Gen. Pharmac, 14(6):603-607.
  • Kendal DA et al. (1982) Comparison of the Antinociceptive Effect of y-Aminobutyric(GABA)Agonists: Evidence for a Cholinergic Involvement, J. Pharmacol. & Therap.220(3):482-487.
  • Hammond EJ and Wilder BJ (1985) Minireview: Gamma-Vinyl GABAGen. Pharmac. 16(5):441-447.
  • Meldrum B and Horton R (1978) Blockade of Epileptic Resonses in the Photosensitive Baboon, Papio papio, by Two Irreversible Inhibitors of GABA-Transaminase, y-Acetylic GABA (4-Aminohex-5-yonic Acid)and y-Vinyl GABA(Amino-hex-5-enoic Acid), Psychopharmacology 59:47-50.
  • Griffin et al. (1993) Peripheral Neuropathy, 3rd ed., (“Griffin 1993”).
  • Presley RW. Novel appriaches to the treatment of neuropathic pain. West J Med. 1992 Nov.; 157(5):564.
  • Simon RP et al. (1989)Clinical Neurology, Appleton & Langel , East Norwalk, CT at p. 72.
  • Cherny NI, et al. [Pharmacotherapy of cancer pain. 3. Adjuvant drugs][Article in German]Schmerz. Mar. 1995; 9(2):55-69.
  • Hegarty A. and Portenoy RK. (1994) Pharmacotherapy of neuropathic pain. Seminars in Neurology, 14,213-224 (Hegarty & Portenoy, 1994).
  • Swerdlow, M. (1984) Anticonvulsant drugs and chronic pain. Clinical Neuropharmacology, 7, 51-82. (Swerdlow, 1984).
  • Kloke (1991) Anti-depressants and anti-convulsants for the treatment of neuropathic pain syndromes in cancer patients. Onkologie 14(1):40-3.
  • Bartusch SL et al. (1996) Clonazepam for the treatment of lancinating phantom limb pain. Clin J Pain. 12(1)59-62.
  • McQuay HJ and Moore RA, (1997) Systematic review of outpatient services for chronic pain control, Chapter 14—Anticonvulsant Drugs, Health Technology Assessment 1997, vol. 1, No. 6: 65-74.
  • Nagahisa A. et al. (1992) Non-specific activity of(±)-CP-96, 345 in models of pain and inflammation, Br. J. Pharmacol. 107:273-275.
  • Winter CA et al. (1962) Carrageenan-induced edema in hind paw of the rat as an assay for anti-inflammatory drugs.Proceedings of the Society for Experimental Biology and Medicine 111:544-547.
  • Kandel, Schwartz & Jessell (1991) Principles of Neural Science, 3rd ed.
  • Dubner R, Hargreaves KM (1989) The neurobiology of pain and its modulation. Clin J Pain 5 Suppl. 2:S1-6.
  • McQuay HJ (1988) Pharmacological treatment of neuralgic and neuropathic pain. Cancer Surveys, 7(1): 141-159).
  • Elliott KJ (1994) Taxonomy and Mechanisms of Neuropathic Pain, Seminars in Neurology 14(3): 195-205.
  • Bennett GJ (1993) An Animal Model of Neuropathic Pain: A Review, Muscle & Nerve 16:1040-1048.
  • Mao J et al. (1993) Patterns of Increased Brain Activity Indicative of Pain in a Rat Model of Peripheral Neuropathy, The Journal of Neuroscience 13(6): 2689-2702.
  • Smith et al. (1994) Increased sensitivity to the antinociceptive activity of (+/−)-baclofen in an animal model of chronic neuropathic, but not chronic inflammatory hyperalgesia. Neuropharmacology, Sep. 1994; 33(9): 1103-8.
  • Merskey, H. & Bogduk, N. (1994) Classification of chronic pain. Descriptions of chronic pain syndromes and definitions of pain terms (2nd ed.) Seattle: IASP Press.
  • Bonica JJ (1990) The Management of Pain, 2nd ed., Lippincott Williams & Wilkins, Philadelphia.
  • Moote CA, The prevention of postoperative pain. Can J Anaesth. Jun. 1994; (6):527-33.
  • Bennett G.J., and Xie Y.K., Pain 1988; 33:87-107.
  • Kim S.H. et al., Pain 1990; 50:355-363.
  • McCaffery M and Pasero P (1999) Pain: Clinical Manual (2nd ed.), Mosby, Inc., St. Louis.
  • Simon RP et al., (1989) Clinical Neurology, Appleton & Lange, East Norwalk, CT, at chapters 3 and 7.
  • Dyck PJ et al. (1987) Diabetic Neuropathy, WB Saunders Co., Philadelphia.
  • Schmidt RE et al. (1981) Experimental Diabetic Autonomic Neuropathy, AJP 103(2): 210-225.
  • Houghton AD et al., Phantom pain: natural history and association with rehabilitation Ann R Coli Surg Engl. Jan. 1994; 76(1):22-5.
  • Volmink J. et al. Treatments for postherpetic neuralgia—a systematic review of randomized controlled trials Fam Pract. Feb. 1996; 13(1):84-91.
  • Bennett GJ. (1994) Hypotheses on the Pathogenesis of Herpes Zoster-associated Pain, Annals of Neurology vol. 35 (Suppl. )S38-S41.
  • Bowsher D. (1997) The management of postherpetic neuralgia, Postgrad Med J 73:623-629.
  • Mamdani, FS (1994) Pharmacologic management of herpes zoster and postherpetic neuralgia, Canadian Family Physician 40:321-332.
  • Wolfe F., Fibromyalgia and myofascial pain syndrome. In Portenoy RK, Kanner RM: Pain management: theory and practice, p. 145-169, Philadelphia, 1996, FA Davis.
  • Carette S., Chronic pain syndromes, Annals of the Rheumatic Diseases 1996; 55: 497-501.
  • Doherty M. and Jones A ABC of rheumatology Fibromyalgia syndrome. BMJ. Feb. 11, 1995; 310(6976): 386-389.
  • Yunus MB (1989) Fibromyalgia syndrome: new research on an old malady, BMJ 298:474-475.
  • Goldenberg DL (1995) Fibromyalgia: why such controversy? Annals of the Rheumatic Diseases 54:3-5.
  • Wade A, Weller, PJ (1994) Handbook of Pharmaceutical Excipients2nd ed., American Pharmaceutical Association, Academy of Pharmaceutical Sciences, Pharmaceutical Society of Great Britain.
  • Neurontin® Product Monograph, revised Feb. 25, 2008.
  • Lyrica® Product Monograph, revised Mar. 3, 2009.
  • Hunter JC. et al., (1997) The effect of novel anti-epileptic drugs in rat experimental models of acute and chronic pain. Eur J Pharmacol. Apr. 18, 1997;324(2-3):153-60.
  • Samkoff LM. et al., (1997) Amelioration of refractory dysesthetic limb pain in multiple sclerosis by gabepentin. Neurology. Jul. 1997; 49(1):304-305.
  • Kim, Yoon C. et al., Glutamic Acid Analogs. The Synthesis of 3-Alkylgutamic Acids and 4-Alkylpyroglutatmic Acids. J. Med. Chem. 1965, 8(4), 509-513.
  • Cahn, R.S. et al., Specification of Molecular Chirality. Angew. Chem. Internat. Edit. vol. 5 (1966) No. 4, 385-583.
  • Moss, G.P., Basic Terminology of Stereochemistry. Pure & Appl. Chem. vol. 68, No. 12, 2193-2222 (1996).
  • Cates, Lindley A. Calculation of Drug Solubilities by Pharmacy Students. American Journal of Pharmaceutical Education. vol. 45, Feb. 1981, 11-13.
  • Schechter, et al. Attempts to Correlate Alterations in Brain GABA Metabolism by GABA-T Inhibitors with their Anticonvulsant Effects. GABA-Biochemistry and CNS Functions. 43-57 (1979).
  • Silverman, Richard B. and Levy, Mark A. Synthesis of (S)-5-Substituted 4-Aminopentanoic Acids: A New Class of y-Aminobutyric Acid Transaminase Inactivators. J. Org. Chem. 1980, 45, 815-818.
  • Mathew, Jacob et al., An Efficent Synthesis of 3-Amino-4-Fluorobutanoic Acid, an Inactivator of GABA Transaminase. Synthetic Communications, 15(5), 377-383 (1985).
  • Silverman, Richard B. et al., Inactivation of y-Aminobutyric Acid Aminotransferase by (Z)-4-Amino-2-fluorobut-2-enoic Acid. Biochemistry 1988, 27, 3285-3289.
  • Prelog, Valdimir et al. Basic Principles of the CIP-System and Proposals for a Revision, Angew. Chem. Int. Ed. Engl. 21 (1982) 567-583.
  • Field, et al. Detection of static and dynamic components of mechanical allodynia in rat models of neuropathic pain: are they signaled by distinct primary sensory neurons? Pain 83 (1990), 303-311.
  • Van, Jon, “Drug Find Worth $700 Million But Chemist Finds It a Tough Sell to Turn Over Project,” Chicago Tribune, Mar. 10, 2008.
  • Silverman, Richard B. From Basic Science to Blockbuster Drug: The Discovery of Lyrica. Angew. Chem. Int. Ed. 2008, 47, 3500-3504.
  • Defeudis, “Central GABA-ergic Systems and Analgesia”, Drug Dev. Res., 3, pp. 1-15, 1983.
  • Harrison, “Modulation of the GABA Receptor Complex by a Steroid Anaesthetic”, Brain Res., 323, pp. 287-292, 1984.
  • Krogsgaard-Larsen, “Heterocyclic Analogues of GABA: Chemistry, Molecular Pharmacology and Therapeutic Areas”, Progress in Medicinal Chemistry, 22, pp. 67-120, 1985.
  • Hernandez, et al., “A Substrate for GABA-ergic Modulation of Dental Pulp Nociceptive Transmission”, J. Dental Res., 65 (Spec. Issue), p. 754, 1986.
  • Andruskiewicz, et al., “A Convenient Synthesis of 3-Alkyl-4-aminobutanoic Acids”, Synthesis, pp. 953-955, Dec. 1989.
  • Andruszkiewicz, et al., “4-Amino-3-Alkylbutanoic Acids as Substrates for Gamma-Aminobutyric Acid Aminotransferase”, Journal of Biological Chemistry, 265 (36), 22288-91, 1990.
  • Silverman, et al., “3-Alkyl-4-aminobutyric Acids: The First Class of Anticonvulsant Agents That Activates L-Glutamic Acid Decarboxylase”, J. Med. Chem., 34, p. 2295-98, 1991.
  • Johnston, “GABAA Agonists as Targets for Drug Development”, Clin. Exp. Pharmacol. and Physiol., 19, pp. 73-78, 1992.
  • Taylor, et al., “Pharmacology of Gabapentin, a Novel Anticonvulsant, In Vitro and in Experimental Animals”, J. Epilepsia, 33 (Suppl. 3), p. 117, 1992.
  • Pfeifer, et al., “A Highly Successful and Novel Model for Treatment of Chronic Painful Diabetic Peripheral Neuropathy”, Diabetes Care, 16 (8), pp. 1103-1115, 1993.
  • Suman-Chauhan, et al., “Characterisation of [3H]Gabapentin Binding to a Novel Site in Rat Brain:Homogenate Binding Studies”, Eur. J. Pharmacol., 244 (3), pp. 293-301, 1993.
  • Taylor, “Mechanism of Action of New Anti-Epileptic Drugs”, Royal Society of Medicine Inter.l Congress and Symposium Series New Trends in Epilepsy Management, 198, p. 13-40, 1993.
  • Taylor, et al., Potent and Stereospecific Anticonvulsant Activity of 3-Isobutyl GABA Relates to in Vitro at a Novel Site Labeled by . . . Epilepsy Res., p. 11-15, 1993.
  • Thurlow, et al., “[3H]Gabapentin May Label a System-L-Like Neutral Amino Acid Carrier in Brain”, European Journal of Pharmacology, 247, pp. 341-345, 1993.
  • Krogsgaard, et al., “GABAA Receptor Agonists, Partial Agonists and Antagonists. Design and Therapeutic Prospects”, J. Med. Chem., 37(16), pp. 2489-2505, 1994.
  • Taylor, “Perspectives on the Pharmacology of Gabapentin (Neurontin) and Potential Mechanism of Action”, Boll. Lega. It. Epii. 86/87, pp. 51-53, 1994.
  • Taylor, “Emerging Perspective on the Mechanism of Action of Gabapentin”, Neurology, 44 (Suppl. 5), pp. S10-S16, 1994.
  • Davies, “Mechanisms of Action of Antiepileptic Drugs”, Seizure, 4, pp. 267-271, 1995.
  • Galer, “Neuropathic pain of Peripheral Origin: Advances in Pharmacologic Treatment”, Neurology, 45 (Suppl. 9), pp. S17-S25, 1995.
  • Radulovic, et al.; “The Preclinical Pharmacology, Pharmacokinetics and Toxicology of Gabapentin”; Drugs of Today, 31 (8), pp. 597-611, 1995.
  • Taylor, “Gabapentin: Mechanisms of Action”, Antiepileptic Drugs (Fourth Edition), pp. 829-841, 1995.
  • Wang, et al., “Pharmacokinetic and Pharmacodynamic Comparison of Two Anticonvulsant Compounds, Gabapentin and Isobutyl GABA”,J. Pharmaceutical Research, 12(Suppl. 9), p. S400 1995.
  • Xiao, et al. Gabapentin Relieves Abnormal Pains in a Rat Model of Painful Peripheral Neuropathy, Soc. For Neuroscience Abstracts, 21, p. 897, 1995.
  • Gilron, et al., “Preemptive Analgesic Effects of Steroid Anesthesia with Aphaxalone in the Rat Formalin Test”, Anesthesiology, 84 (3), pp. 572-579, 1996.
  • Hill, “Meeting Highlights Central & Peripheral Nervous Systems ( Butt 8th World Pain Congress)”, Expert Opinion Invest. Drugs, 5(11), pp. 1549-1562, 1996.
  • Mathew, et al., “Gabapentin in Migraine Prophylaxis: A Preliminary Open Label Study”, Neurology, 46 (Suppl. 2), p. A169, 1996.
  • Schachter, et al., “Treatment of Central Pain with Gabapentin: Case reports”, J. of Epilepsy, 9(3), pp. 223-226, 1996.
  • Shimoyama, et al., “Spinal Gabapentin is Antinoceptive in the Rat Formalin Test”, Society for Neuroscience Abstracts, 22, p. 1371, 1996.
  • Singh, et al., “The Antiepileptic Agent Gabapentin Posseses Anxiolytic-Like and Antinociceptive Actions that are Reversed by D-Serine”, Psychopharmacology, 127, pp. 1-9, 1996.
  • Stacey, et al., “Gabapentin and Neuropathic Pain States: A Case Series Report”, Regional Anesthesia, 21 (Suppl. 2), p. 65, 1996.
  • Vinik, et al., “Recent Advances in the Diagnosis and Treatment of Diabetic Neuropathy”, Endocrinologist, 6 (6), pp. 443-461, 1996.
  • Xiao, et al., “Gabapentin Has an Antinociceptive Effect Mediated via a Spinal Site of Action in a Rat Model of Painful Peripheral Neuropathy”, Analgesia, 2, pp. 267-273, 1996.
  • Field, et al., “Gabapentin (Neurontin) and S-(+)-3-Isobutylgaba Represent a Novel Class of Selective Antihyperalgesic Agents”, Br. J. Parmacol., 121, pp. 1513-1522, 1997.
  • Field, et al., “Evaluation of Gabapentin and S-(+)-3-Isobutylgaba in a Rat Model of Postoperative Pain”, J. Pharmacol. Exp. Ther., 282 (3), pp. 1242-1246, 1997.
  • Bryans, et al., “Gabapentin SAR-Towards Novel Treatments for Pain”, Abstracts of Papers American Chemical Society, 216(1), p. MEDI 207, 1998.
  • Bryans, et al., “Identification of Novel Ligands for the Gabapentin Binding Site on the α2σ Subunit of a Calcium Channel and . . . ”, J. Med. Chem., 41, p1838-45, 1998.
  • Partridge, et al, “characterization of the Effects of Gabapentin and 3-Isobutyl-y-Aminobutyric Acid on Substance P-induced. . .”, Anesthesiology, 88 (1), p196-205, 1998.
  • Taylor, et al., “A Summary of Mechanistic Hypothesis of Gabapentin Pharmacology”, Epilepsy Research, 29, pp. 233-249, 1998.
  • Field, et al., “Gabapentin and Pregabalin, but not Morphine and Amitriptyline, Block Both Static and Dynamic Components of Mechanical . . . ”, Pain (Neth.), 80, p391-98, Mar. 1999.
  • Field, et al., “The Gabapentin Analogue 3-Methyl-Gabapentin Blocks Both Static and Dynamic Components of Mechanical Allodynia in . . . ”, British J. of Pharm., 128, p235P, 1999.
  • Field, et al., “Detection of Static and Dynamic Components of Mechanical Allodynia in Rat Models of NeuropathicPain: Are They Signalled by . . . ”, 83, p303-11, 1999.
  • Blakemore, et al., “Gabapentin SAR: Toward Novel Treatment for Pain”, Abstracts of Papers Americna Chemical Society, 220 (Part 1), p. MEDI 239, 2000.
  • Field, et al., “Futher Evidence for the Role of the α2σ Subunit of Voltage Dependent Calcium Channels in Models of NeuropathicPain”, Br. J. of Pharm., 131 (2), p282-86, 2000.
  • Laird, et al., “Use of Gabapentin in the Treatment of Neuropathic Pain”, Annals of Pharmacotherapy, 34, pp. 802-807, 2000.
  • Buirkle, et al., “Pregabalin Inhibits Mechanical Hyperalgesia Origination in the Musculoskeletal System”, J. Society for Neuroscience Abstracts, 27(1), p. 1332, 2001.
  • Taylor, et al., “Pregabalin Inhibits Multiple Endpoints of Carrageenan Induced Pain but Not Inflammation in Rats”, J. Society for Neuroscience Abstracts, 27(2), p. 1897, 2001.
  • Rose, et al., “Gabapentin: Pharmacology and Its Use in Pain Management”, Anaesthesia, 57, pp. 451-462, 2002.
  • Bellioti, et al., “Structure-Activity Relationships of Pregabalin and Analogues that Target the α2σ Protein”, J. Med. Chem, 48 (7), pp. 2294-2307, 2005.
  • Bian, et al., “Calcium Channel Alpha2-Delta Type 1 Subunit is the Major Binding Protein for Pregabalin in Neocortex, Hippocampus, . . . ”, Brain Research, 1075, p. 68-80, 2006.
  • Rogawski, et al., “Calcium α2-σ Subunit, A New Antiepileptic Drug Target”, Epilepsy Res., 69(3), pp. 183-272, 2006.
  • Vartanian, et al., “Activity Profile of Pregabalin in Rodent Models of Epilepsy and Ataxia”, Epilepsy Res., 68, pp. 189-205, 2006.
  • Taylor, et al, “Pharmacology and Mechanism of Action of Pregablin: The Calcium Channel α2-σ (Alpha2-Delta) Subunit as a Target . . . ”, Epilepsy Res., 73, p. 137-50, 2007.
  • Yuen et al., Enantioselective Synthesis of PD144723: A Potent Stereospecific Anticonvulsant, Bioorg. Medicinal Chemistry Letters, 4 (6), pp. 823-826, 1994.
  • Lyrica U.S. Physician Prescribing Information.
  • Neurontin U.S. Physician Prescribing Information.
  • EP 0934061 File History (including Opposition Document)s.
  • CA 2,255,652 File History (including Application for Reissue filed Dec. 20, 2005).
  • Apr. 14, 2005 Declaration of Charles Taylor submitted in Japanese Application No. JP507062/98.
  • Translation of Written Opinion of ANVISA refusing to consent to grant of patent based on allowed Brazillian Patent Application No. P19710536-8.
Patent History
Patent number: RE41920
Type: Grant
Filed: Jul 16, 1997
Date of Patent: Nov 9, 2010
Assignee: Warner-Lambert Company LLC (New York, NY)
Inventor: Lakhbir Singh (Cambridgeshire)
Primary Examiner: Sreeni Padmanabhan
Assistant Examiner: Craig Ricci
Attorney: Connolly Bove Lodge & Hutz LLP
Application Number: 11/983,750
Classifications
Current U.S. Class: Nitrogen Other Than As Nitro Or Nitroso Nonionically Bonded (514/561)
International Classification: A61K 31/195 (20060101);