US20110144188A1 - Salts of trimebutine and n-desmethyl trimebutine - Google Patents

Salts of trimebutine and n-desmethyl trimebutine Download PDF

Info

Publication number
US20110144188A1
US20110144188A1 US12/303,082 US30308207A US2011144188A1 US 20110144188 A1 US20110144188 A1 US 20110144188A1 US 30308207 A US30308207 A US 30308207A US 2011144188 A1 US2011144188 A1 US 2011144188A1
Authority
US
United States
Prior art keywords
trimebutine
compound
nitroargininate
compound according
desmethyltrimebutine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/303,082
Inventor
John L. Wallace
Giuseppe Cirino
Vincenzo Santagada
Giuseppe Caliendo
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GICARE PHARMA Inc
Original Assignee
Antibe Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Antibe Therapeutics Inc filed Critical Antibe Therapeutics Inc
Priority to US12/303,082 priority Critical patent/US20110144188A1/en
Assigned to ANTIBE THERAPEUTICS INC. reassignment ANTIBE THERAPEUTICS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WALLACE, JOHN L., CALIENDO, GIUSEPPE, CIRINO, GIUSEPPE, SANTAGADA, VINCENZO
Publication of US20110144188A1 publication Critical patent/US20110144188A1/en
Assigned to GICARE PHARMA INC. reassignment GICARE PHARMA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ANTIBE HOLDINGS INC.
Assigned to ANTIBE HOLDINGS INC. reassignment ANTIBE HOLDINGS INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: ANTIBE THERAPEUTICS INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C219/00Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton
    • C07C219/02Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C219/04Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C219/14Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having at least one of the hydroxy groups esterified by a carboxylic acid having the esterifying carboxyl group bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C279/00Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups
    • C07C279/30Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of guanidine groups bound to nitro or nitroso groups
    • C07C279/32N-nitroguanidines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C219/00Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton
    • C07C219/02Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C219/00Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton
    • C07C219/02Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C219/20Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being unsaturated
    • C07C219/22Compounds containing amino and esterified hydroxy groups bound to the same carbon skeleton having esterified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being unsaturated and containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/60Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton with the carbon atom of at least one of the carboxyl groups bound to nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C327/00Thiocarboxylic acids
    • C07C327/38Amides of thiocarboxylic acids
    • C07C327/48Amides of thiocarboxylic acids having carbon atoms of thiocarboxamide groups bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D339/00Heterocyclic compounds containing rings having two sulfur atoms as the only ring hetero atoms
    • C07D339/02Five-membered rings
    • C07D339/04Five-membered rings having the hetero atoms in positions 1 and 2, e.g. lipoic acid

Definitions

  • the present invention relates to unique salts of trimebutine and N-monodesmethyl trimebutine, and their corresponding stereoisomers, having improved analgesic properties useful in the treatment of visceral pain in general and more particularly in the treatment of conditions characterized by abdominal pain, for example, in patients with intestinal diseases such as inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS), diabetic gastroparesis, and dyspepsia.
  • intestinal diseases such as inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS), diabetic gastroparesis, and dyspepsia.
  • Trimebutine[3,4,5-trimethoxybenzoic acid 2-(dimethylamino)-2-phenylbutylester and its maleate salt] has been used in many countries since 1969 for the treatment of functional bowel disorders, including IBS.
  • the efficacy of trimebutine to relieve abdominal pain has been demonstrated in various clinical studies (see, for example, Ghidini et al (1986) Single drug treatment for irritable colon: Rociverine versus trimebutine maleate. Curr Ther Res 39: 541-548).
  • Trimebutine has proved to be effective in the treatment of both acute and chronic abdominal pain in patients with functional bowel disorders, especially IBS, at doses ranging from 300 to 600 mg/day. It is also effective in children presenting with abdominal pain.
  • trimebutine has also been shown to decrease reflexes induced by distension of the gut lumen in animals to modulate visceral sensitivity.
  • Nitric oxide (NO) been recently been shown to exert many anti-inflammatory effects, including reduction of leukocyte adherence to the vascular endothelium (Gauthier et at (1994) Nitric oxide attenuates leukocyte-endothelial interaction via P-selectin in splanchnic ischemia-reperfusion. Am J Physiol 267: G562-G568) and suppression of production of various chemotactic factors (Walford and Loscalzo (2003) Nitric oxide in vascular biology. J Thromb Haemost 1: 2112-2118). Further, incorporation of an NO-releasing moiety into certain drugs such as NSAIDs, acetaminophen and ursodexycholic acid has been shown to enhance activities of these drugs and reduce toxicity relative to the parent drug.
  • Hydrogen sulfide is another type of gaseous mediator that may exert anti-inflammatory effects. Recently it has been shown that H 2 S releasing agents exhibit analgesic activity in models of visceral pain (Distrutti et at (2005) Evidence that hydrogen sulfide exerts antinociceptive effects in the gastrointestinal tract by activating K ATP channels. J Pharmacol Exp Ther 316: 325-335. Further, H 2 S has been shown to be a smooth muscle relaxant in intestinal tissues (see Teague, B. et al. (2002) The Smooth Muscle Relaxant effect of Hydrogen Sulfide in Vitro: Evidence for a Physiological Role to Control Intestinal Contractility. Br. J. Pharmacol. 137: 139-145.
  • trimebutine is significantly enhanced when salts of trimebutine or N-monodesmethyl trimebutine and their corresponding stereoisomers are formed with various NO-releasing, H 2 S-releasing or combined NO- and H 2 S-releasing moieties.
  • administration of these NO-releasing, H 2 S-releasing or combined NO- and H 2 S-releasing salts of trimebutine and N-monodesmethyl trimebutine results in improved analgesic properties when compared to trimebutine (trimebutine maleate) or its metabolite N-monodesmethyl trimebutine alone and when compared to the NO-releasing, H 2 S-releasing or combined NO- and H 2 S-releasing moiety alone.
  • These salts are particularly useful in the treatment of conditions characterized by abdominal pain such as such as irritable bowel syndrome, inflammatory bowel disease, diabetic gastroparesis, dyspepsia and the like.
  • salts of trimebutine (TMB) and its active metabolite N-desmethyl trimebutine (Nor-TMB) and their corresponding stereoisomers, (R)-TBM, (S)-TMB, (R)-Nor-TBM and (S)-Nor-TBM, are provided, said salts being formed using NO-releasing, H 2 S-releasing or combined NO- and H 2 S-releasing moieties.
  • the TMB and Nor-TMB salts of the present invention are superior to TMB and Nor-TMB alone and to an NO-releasing, H 2 S-releasing or combined NO- and H 2 S-releasing moiety alone in reducing visceral pain associated with colorectal distension.
  • the NO-releasing, H 2 S-releasing or combined NO- and H 2 S-releasing moieties alone did not appear to have any significant effects on visceral pain associated with colorectal distension when administered alone.
  • the salts of the present invention are useful in alleviating pain associated with any disorder of the digestive system that is associated with abdominal pain.
  • salts of the invention have the following general formula:
  • X is selected from the group consisting of:
  • any non-toxic, effective NO-releasing, H 2 S-releasing or combined NO- and H 2 S-releasing moiety can be used in the present invention.
  • Preferred compounds are those of the following formulae:
  • novel combined NO- and H 2 S-releasing moieties are provided having the general formula, nitroarginine-R, preferably:
  • R is an H 2 S-releasing moiety.
  • R is selected from the group consisting of 5-p-hydroxyphenyl-1,2-dithione-3-thione, cysteine, and 4-(thiocarbamoyl)benzoic acid.
  • a method for treating visceral pain in a subject in need of such treatment comprising administering to the subject a visceral pain relieving amount of a compound of the present invention.
  • the visceral pain is abdominal pain.
  • the abdominal pain is due to intestinal diseases such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), diabetic gastroparesis, and dyspepsia.
  • FIG. 1( a ) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle and trimebutine maleate.
  • FIG. 1( b ) shows the colorectal pressure (mmHg) in a rat model of visceral pain perception using vehicle and trimebutine maleate alone.
  • FIG. 2( a ) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle and nitroarginine alone.
  • FIG. 2( b ) shows the colorectal pressure (mmHg) in a rat model of visceral pain perception using vehicle and nitroarginine alone.
  • FIG. 3( a ) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle and trimebutine nitroargininate (salt I).
  • FIG. 3( b ) shows the colorectal pressure (mmHg) in a rat model of visceral pain perception using vehicle and trimebutine nitroargininate (salt I).
  • FIG. 4( a ) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle or trimebutine nitroargininate (salt I), with or without pretreatment with L-NAME.
  • FIG. 4( b ) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle or trimebutine nitroargininate (salt I), with or without pretreatment with methylene blue.
  • FIG. 5( a ) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle, trimebutine maleate and trimebutine thiocarbamoylbenzoate (salt III).
  • FIG. 5( b ) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle and thiocarbamoylbenzoate (TBZ) alone.
  • FIG. 6 is a bar graph showing H 2 S generation of 4-(thiocarbamoyl)benzoic acid (TBZ) and 5-(4-amino-phenyl)-[1,2]dithiole-3-thione (ADT-OH).
  • the compounds of the present invention contain two active moieties, (1) either TMB or Nor-TMB, or their stereoisomers, and (2) an NO-releasing moiety, an H 2 S-releasing moiety, or combined NO- and H 2 S-releasing moieties.
  • the salts of the present invention can be made using known starting materials and reagents.
  • Compounds of the present invention may be utilized for the treatment of visceral pain, such as abdominal pain, associated with various diseases, including, but not limited to, Crohn's disease, ulcerative colitis, irritable bowel syndrome, infectious colitis (e.g., pseudomembranous colitis such as Clostridium difficile colitis, salmonella enteritis, shigella infections, yersiniosis, cryptospiridiosis, microspridial infections, and viral infections), radiation-induced colitis, colitis in the immunocompromised host, diabetic gastroparesis and dyspepsia.
  • infectious colitis e.g., pseudomembranous colitis such as Clostridium difficile colitis, salmonella enteritis, shigella infections, yersiniosis, cryptospiridiosis, microspridial infections, and viral infections
  • radiation-induced colitis e.g., radiation-induced colitis in the immunocompromised host, diabetic gastroparesis and dyspepsia
  • subjects may be administered compounds of the present invention at any suitable therapeutically effective and safe dosage, as may be readily determined within the skill of the art.
  • These compounds are, most desirably, administered in dosages ranging from about 1 to about 2000 mg per day, in a single or divided doses, although variations will necessarily occur depending upon the weight and condition of the subject being treated and the particular route of administration chosen.
  • a dosage level that is in the range of about 0.1 to about 100 mg/kg, preferably between about 5 and 90 mg/kg, and more preferably between about 5 and 50 mg/kg, is most desirable.
  • Variations may nevertheless occur depending upon the weight and conditions of the persons being treated and their individual responses to said medicament, as well as on the type of pharmaceutical formulation chosen and the time period and interval during which such administration is carried out.
  • dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effects, provided that such large doses are first divided into several small doses for administration throughout the day.
  • the compounds of the present invention can be administered in the form of any pharmaceutical formulation, the nature of which will depend upon the route of administration.
  • These pharmaceutical compositions can be prepared by conventional methods, using compatible, pharmaceutically acceptable excipients or vehicles. Examples of such compositions include capsules, tablets, transdermal patches, lozenges, troches, sprays, syrups, powders, granulates, gels, elixirs, suppositories, and the like, for the preparation of extemporaneous solutions, injectable preparations, rectal, nasal, ocular, vaginal etc.
  • a preferred route of administration is the oral and rectal route.
  • tablets containing various excipients such as microcrystalline cellulose, sodium citrate, calcium carbonate, dicalcium phosphate and glycine may be employed along with various disintegrants such as starch (preferably corn, potato or tapioca starch), alginic acid and certain complex silicates, together with granulation binders like polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • disintegrants such as starch (preferably corn, potato or tapioca starch), alginic acid and certain complex silicates, together with granulation binders like polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc can be used for tabletting purposes.
  • Solid compositions of similar type may also be employed as fillers in gelatin capsules; preferred materials in this connection also include lactose or milk sugar, as well as high molecular weight polyethylene glycols.
  • the active ingredient may be combined with sweetening or flavoring agents, coloring matter and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin and various combinations thereof.
  • the dosage form can be designed for immediate release, controlled release, extended release, delayed release or targeted delayed release.
  • the definitions of these terms are known to those skilled in the art.
  • the dosage form release profile can be effected by a polymeric mixture composition, a coated matrix composition, a multiparticulate composition, a coated multiparticulate composition, an ion-exchange resin-based composition, an osmosis-based composition, or a biodegradable polymeric composition. Without wishing to be bound by theory, it is believed that the release may be effected through favorable diffusion, dissolution, erosion, ion-exchange; osmosis or combinations thereof.
  • a solution of an active salt in either sesame or peanut oil or in aqueous propylene glycol can be employed.
  • the aqueous solutions should be suitably buffered (preferably pH greater than 8), if necessary, and the liquid diluent first rendered isotonic.
  • the aqueous solutions are suitable for intravenous injection purposes. The preparation of all these solutions under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • the crude intermediate a was treated with a solution of trifluoroacetic acid in dichloromethane (40% TFA in DCM). After 1 h the solvent was removed to obtain H-Cys-Arg(NO 2 )—OH.TFA as a crude residue which was precipitated with diethyl ether; the obtained solid was dissolved in water and 1N NaOH was slowly added to obtain 2-(2-amino-3-mercapto-propionylamino)-5-nitroguanidino-pentanoic acid (b) as a white solid which was recovered by filtration.
  • the crude product 5 was dissolved in 22 mL of anhydrous THF and 2.64 mL of triethyl amine were added. After stirring for 30 minutes, the mixture was filtered through celite and the solid washed with 100 mL of anhydrous diethyl ether. Concentration of the solvents using a rotary evaporator afforded 2.49 g of 6 as a blue oil (77% yield).
  • the crude intermediate a was treated with a solution of trifluoroacetic acid in dichloromethane (40% TFA in DCM). After 1 h the solvent was removed to obtain H-Cys-Arg(NO 2 )-OH.TFA as a crude residue which was precipitated with diethyl ether; the obtained solid was dissolved in water and 1N NaOH was slowly added to obtain 2-(2-amino-3-mercapto-propionylamino)-5-nitroguanidino-pentanoic acid (b) as a white solid which was recovered by filtration.
  • Rats male, Wistar, 200-250 g, obtained from Charles River, Monza, Italy
  • Rats were housed in plastic cages and maintained under controlled conditions with 12-hours light/dark cycle with lights on at 7.00 AM. Tap water and standard laboratory chow were freely available.
  • rats were individually trained by spending 2-3 hours per day in a plexiglass cage for 2-3 days. It allowed them to adjust to a movement-restriction environment. Food was withheld for 12 hours before colorectal distension (CRD) recording were performed.
  • CCD colorectal distension
  • rats were sedated with ether inhalation and a 2 cm long latex balloon was inserted intrarectally 2 cm from the anal verge and fixed at the base of the tail.
  • the balloon was connected via a double-barreled cannula to a pressure transducer to continuously monitoring the rectal pressure by a computer (PowerLab PC, A.D. Instruments, Milford, Mass., USA) and to a syringe for inflation/deflation of the balloon.
  • the rats were then housed in a small cage (20 ⁇ 8 ⁇ 8 cm) on an elevated PlexiglasTM platform and allowed to wake up and adapt for 1 hour. After recovery from sedation, animals underwent the CRD procedure and behavioral responses were tested. The night before the experiments, the balloons were inflated and left overnight so the latex stretched and the balloons became compliant.
  • CRD CRD of 20 seconds, performed every 5 minutes, was applied in increment of 0.4 ml starting from 0.4 ml up to 1.6 ml water. To achieve an accurate measurement of the colonic parameters and perception, the distensions were repeated twice for each intensity and data for each animal were averaged for analysis. Each animal underwent a double set of CRD. Twenty minutes after the first sequence of CRD (0.4 mL-1.6 ml water), drugs were administered intraperitoneally (i.p.) and a second set of CRD was performed. Behavioral responses during the first and the second set of CRD were assessed and compared.
  • AWR abdominal withdrawal reflex
  • the AWR is an involuntary motor reflex similar to the visceromotor reflex, but it has the great advantage that, in contrast to the latter, it does not require abdominal surgery to implant recording electrodes and wires in the abdominal muscle wall which may cause additional sensitization (see Ness, T. J. and Gebhart, G. F. (1990) Pain 41:167-234, incorporated herein by reference).
  • AWR AWR
  • grade 0 corresponds to no behavioral response to CRD
  • grade 1 corresponds to brief head movement at the onset of the stimulus followed by immobility
  • grade 2 corresponds to a mild contraction of abdominal muscles although the rats does not lift the abdomen off the platform
  • grade 3 corresponds to a strong contraction of the abdominal muscles with the lifting of the abdomen off the platform
  • grade 4 corresponds to a severe contraction of the abdominal muscle manifested by body arching and the lifting of the abdomen and of the pelvic structures and scrotum.
  • trimebutine maleate, nitroarginine and trimebutine nitroargininate were determined using a total of 15 fasting rats.
  • trimebutine maleate, nitroarginine and trimebutine nitroargininate could reduce pain induced by CRD, after the first sequence of CRD (vehicle-treated), 5 rats were treated with trimebutine maleate at a dose of 10 mg/kg i.p., nitroarginine at a dose of 6 mg/kg or trimebutine nitroargininate at the dose of 16 mg/kg i.p., after which a second set of CRD was repeated. Results from these experiments are shown in FIGS. 1( a ), 2 ( a ) and 3 ( a ).
  • FIGS. 1( a ), 2 ( a ) and 3 ( a ) show that trimebutine nitroargininate is more effective than either trimebutine maleate or nitroarginine in reducing visceral pain in response to colorectal distension.
  • none of the compounds were particularly effective in reducing intrarectal pressure, as shown in FIGS. 1( b ), 2 ( b ) and 3 ( b ).
  • FIGS. 1( a ), 2 ( a ) and 3 ( a ) show that trimebutine nitroargininate is more effective than either trimebutine maleate or nitroarginine in reducing visceral pain in response to colorectal distension.
  • none of the compounds were particularly effective in reducing intrarectal pressure, as shown in FIGS. 1( b ), 2 ( b ) and 3 ( b ).
  • trimebutine nitroargininate is useful in treating abdominal pain associated with various inflammatory conditions of the alimentary tract, as well as functional gastrointestinal disorders such as irritable bowel syndrome, dyspepsia, etc., that are characterized by increased visceral nociception (with or without accompanying inflammation).
  • FIGS. 5( a ) and 5 ( b ) show that trimebutine thiocarbamoylbenzoate is more effective than either trimebutine maleate or thiocarbamoylbenzoate in reducing visceral pain in response to colorectal distension.
  • trimebutine thiocarbamoylbenzoate is useful in treating abdominal pain associated with various inflammatory conditions of the alimentary tract, as well as functional gastrointestinal disorders such as irritable bowel syndrome, dyspepsia, etc., that are characterized by increased visceral nociception (with or without accompanying inflammation).

Abstract

Unique salts of trimebutine and N-monodesmethyl trimebutine, and their corresponding stereoisomers, having improved analgesic properties useful in the treatment of visceral pain are provided. The salts of the present invention are particularly useful in the treatment of conditions characterized by abdominal pain, such as inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS), diabetic gastroparesis, and dyspepsia.

Description

  • This application claims the benefit of U.S. provisional patent application No. 60/804,067, filed Jun. 6, 2006.
  • FIELD OF INVENTION
  • The present invention relates to unique salts of trimebutine and N-monodesmethyl trimebutine, and their corresponding stereoisomers, having improved analgesic properties useful in the treatment of visceral pain in general and more particularly in the treatment of conditions characterized by abdominal pain, for example, in patients with intestinal diseases such as inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS), diabetic gastroparesis, and dyspepsia.
  • BACKGROUND OF THE INVENTION
  • Trimebutine[3,4,5-trimethoxybenzoic acid 2-(dimethylamino)-2-phenylbutylester and its maleate salt] has been used in many countries since 1969 for the treatment of functional bowel disorders, including IBS. The efficacy of trimebutine to relieve abdominal pain has been demonstrated in various clinical studies (see, for example, Ghidini et al (1986) Single drug treatment for irritable colon: Rociverine versus trimebutine maleate. Curr Ther Res 39: 541-548). Trimebutine has proved to be effective in the treatment of both acute and chronic abdominal pain in patients with functional bowel disorders, especially IBS, at doses ranging from 300 to 600 mg/day. It is also effective in children presenting with abdominal pain.
  • It is thought that the actions of trimebutine on the gastrointestinal tract are mediated in part via (i) an agonist effect on peripheral mu, kappa and delta opiate receptors and (ii) release of gastrointestinal peptides such as motilin and modulation of the release of other peptides, including vasoactive intestinal peptide, gastrin and glucagon. Further, trimebutine accelerates gastric emptying, induces premature phase III of the migrating motor complex in the intestine and modulates the contractile activity of the colon. Recently, trimebutine has also been shown to decrease reflexes induced by distension of the gut lumen in animals to modulate visceral sensitivity.
  • Nitric oxide (NO) been recently been shown to exert many anti-inflammatory effects, including reduction of leukocyte adherence to the vascular endothelium (Gauthier et at (1994) Nitric oxide attenuates leukocyte-endothelial interaction via P-selectin in splanchnic ischemia-reperfusion. Am J Physiol 267: G562-G568) and suppression of production of various chemotactic factors (Walford and Loscalzo (2003) Nitric oxide in vascular biology. J Thromb Haemost 1: 2112-2118). Further, incorporation of an NO-releasing moiety into certain drugs such as NSAIDs, acetaminophen and ursodexycholic acid has been shown to enhance activities of these drugs and reduce toxicity relative to the parent drug.
  • Hydrogen sulfide (H2S) is another type of gaseous mediator that may exert anti-inflammatory effects Recently it has been shown that H2S releasing agents exhibit analgesic activity in models of visceral pain (Distrutti et at (2005) Evidence that hydrogen sulfide exerts antinociceptive effects in the gastrointestinal tract by activating KATP channels. J Pharmacol Exp Ther 316: 325-335. Further, H2S has been shown to be a smooth muscle relaxant in intestinal tissues (see Teague, B. et al. (2002) The Smooth Muscle Relaxant effect of Hydrogen Sulfide in Vitro: Evidence for a Physiological Role to Control Intestinal Contractility. Br. J. Pharmacol. 137: 139-145.
  • The inventors have shown in the present application that the activity of trimebutine is significantly enhanced when salts of trimebutine or N-monodesmethyl trimebutine and their corresponding stereoisomers are formed with various NO-releasing, H2S-releasing or combined NO- and H2S-releasing moieties. In particular, administration of these NO-releasing, H2S-releasing or combined NO- and H2S-releasing salts of trimebutine and N-monodesmethyl trimebutine results in improved analgesic properties when compared to trimebutine (trimebutine maleate) or its metabolite N-monodesmethyl trimebutine alone and when compared to the NO-releasing, H2S-releasing or combined NO- and H2S-releasing moiety alone. These salts are particularly useful in the treatment of conditions characterized by abdominal pain such as such as irritable bowel syndrome, inflammatory bowel disease, diabetic gastroparesis, dyspepsia and the like.
  • SUMMARY OF THE INVENTION
  • In general, salts of trimebutine (TMB) and its active metabolite N-desmethyl trimebutine (Nor-TMB) and their corresponding stereoisomers, (R)-TBM, (S)-TMB, (R)-Nor-TBM and (S)-Nor-TBM, are provided, said salts being formed using NO-releasing, H2S-releasing or combined NO- and H2S-releasing moieties. By forming a salt with an NO-releasing, H2S-releasing or combined NO- and H2S-releasing moiety, the anti-nociceptive effect of TMB and Nor-TMB was surprisingly enhanced.
  • More particularly, the TMB and Nor-TMB salts of the present invention are superior to TMB and Nor-TMB alone and to an NO-releasing, H2S-releasing or combined NO- and H2S-releasing moiety alone in reducing visceral pain associated with colorectal distension. The NO-releasing, H2S-releasing or combined NO- and H2S-releasing moieties alone did not appear to have any significant effects on visceral pain associated with colorectal distension when administered alone. Thus, in one aspect of the invention, the salts of the present invention are useful in alleviating pain associated with any disorder of the digestive system that is associated with abdominal pain.
  • Broadly stated, salts of the invention have the following general formula:

  • A+·X  (formula I)
  • where:
    • A is
  • Figure US20110144188A1-20110616-C00001
    • and their corresponding stereoisomers;
    • and X is a NO-releasing, H2S-releasing or combined NO- and H2S-releasing moiety.
  • In a preferred embodiment, X is selected from the group consisting of:
  • Figure US20110144188A1-20110616-C00002
  • It is understood that any non-toxic, effective NO-releasing, H2S-releasing or combined NO- and H2S-releasing moiety can be used in the present invention.
  • Preferred compounds are those of the following formulae:
  • Figure US20110144188A1-20110616-C00003
    Figure US20110144188A1-20110616-C00004
    Figure US20110144188A1-20110616-C00005
  • Further, novel combined NO- and H2S-releasing moieties are provided having the general formula, nitroarginine-R, preferably:
  • Figure US20110144188A1-20110616-C00006
  • wherein R is an H2S-releasing moiety. In a preferred embodiment, R is selected from the group consisting of 5-p-hydroxyphenyl-1,2-dithione-3-thione, cysteine, and 4-(thiocarbamoyl)benzoic acid.
  • Further, a method for treating visceral pain in a subject in need of such treatment is provided comprising administering to the subject a visceral pain relieving amount of a compound of the present invention. In one embodiment, the visceral pain is abdominal pain. In another embodiment, the abdominal pain is due to intestinal diseases such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), diabetic gastroparesis, and dyspepsia.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1( a) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle and trimebutine maleate.
  • FIG. 1( b) shows the colorectal pressure (mmHg) in a rat model of visceral pain perception using vehicle and trimebutine maleate alone.
  • FIG. 2( a) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle and nitroarginine alone.
  • FIG. 2( b) shows the colorectal pressure (mmHg) in a rat model of visceral pain perception using vehicle and nitroarginine alone.
  • FIG. 3( a) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle and trimebutine nitroargininate (salt I).
  • FIG. 3( b) shows the colorectal pressure (mmHg) in a rat model of visceral pain perception using vehicle and trimebutine nitroargininate (salt I).
  • FIG. 4( a) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle or trimebutine nitroargininate (salt I), with or without pretreatment with L-NAME.
  • FIG. 4( b) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle or trimebutine nitroargininate (salt I), with or without pretreatment with methylene blue.
  • FIG. 5( a) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle, trimebutine maleate and trimebutine thiocarbamoylbenzoate (salt III).
  • FIG. 5( b) shows the perception score (AWR Score) in a rat model of visceral pain perception using vehicle and thiocarbamoylbenzoate (TBZ) alone.
  • FIG. 6 is a bar graph showing H2S generation of 4-(thiocarbamoyl)benzoic acid (TBZ) and 5-(4-amino-phenyl)-[1,2]dithiole-3-thione (ADT-OH).
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
  • The invention will now be described with respect to preferred embodiments described herein. It should be appreciated however that these embodiments are for the purpose of illustrating the invention, and are not to be construed as limiting the scope of the invention as defined by the claims.
  • The compounds of the present invention contain two active moieties, (1) either TMB or Nor-TMB, or their stereoisomers, and (2) an NO-releasing moiety, an H2S-releasing moiety, or combined NO- and H2S-releasing moieties. In many instances, the salts of the present invention can be made using known starting materials and reagents.
  • Compounds of the present invention may be utilized for the treatment of visceral pain, such as abdominal pain, associated with various diseases, including, but not limited to, Crohn's disease, ulcerative colitis, irritable bowel syndrome, infectious colitis (e.g., pseudomembranous colitis such as Clostridium difficile colitis, salmonella enteritis, shigella infections, yersiniosis, cryptospiridiosis, microspridial infections, and viral infections), radiation-induced colitis, colitis in the immunocompromised host, diabetic gastroparesis and dyspepsia.
  • Depending on the specific condition or disease state to be treated, subjects may be administered compounds of the present invention at any suitable therapeutically effective and safe dosage, as may be readily determined within the skill of the art. These compounds are, most desirably, administered in dosages ranging from about 1 to about 2000 mg per day, in a single or divided doses, although variations will necessarily occur depending upon the weight and condition of the subject being treated and the particular route of administration chosen. However, a dosage level that is in the range of about 0.1 to about 100 mg/kg, preferably between about 5 and 90 mg/kg, and more preferably between about 5 and 50 mg/kg, is most desirable. Variations may nevertheless occur depending upon the weight and conditions of the persons being treated and their individual responses to said medicament, as well as on the type of pharmaceutical formulation chosen and the time period and interval during which such administration is carried out. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effects, provided that such large doses are first divided into several small doses for administration throughout the day.
  • The compounds of the present invention can be administered in the form of any pharmaceutical formulation, the nature of which will depend upon the route of administration. These pharmaceutical compositions can be prepared by conventional methods, using compatible, pharmaceutically acceptable excipients or vehicles. Examples of such compositions include capsules, tablets, transdermal patches, lozenges, troches, sprays, syrups, powders, granulates, gels, elixirs, suppositories, and the like, for the preparation of extemporaneous solutions, injectable preparations, rectal, nasal, ocular, vaginal etc. A preferred route of administration is the oral and rectal route.
  • For oral administration, tablets containing various excipients such as microcrystalline cellulose, sodium citrate, calcium carbonate, dicalcium phosphate and glycine may be employed along with various disintegrants such as starch (preferably corn, potato or tapioca starch), alginic acid and certain complex silicates, together with granulation binders like polyvinylpyrrolidone, sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc can be used for tabletting purposes. Solid compositions of similar type may also be employed as fillers in gelatin capsules; preferred materials in this connection also include lactose or milk sugar, as well as high molecular weight polyethylene glycols. When aqueous suspensions and/or elixirs are desired for oral administration the active ingredient may be combined with sweetening or flavoring agents, coloring matter and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin and various combinations thereof.
  • The dosage form can be designed for immediate release, controlled release, extended release, delayed release or targeted delayed release. The definitions of these terms are known to those skilled in the art. Furthermore, the dosage form release profile can be effected by a polymeric mixture composition, a coated matrix composition, a multiparticulate composition, a coated multiparticulate composition, an ion-exchange resin-based composition, an osmosis-based composition, or a biodegradable polymeric composition. Without wishing to be bound by theory, it is believed that the release may be effected through favorable diffusion, dissolution, erosion, ion-exchange; osmosis or combinations thereof.
  • For parenteral administration, a solution of an active salt in either sesame or peanut oil or in aqueous propylene glycol can be employed. The aqueous solutions should be suitably buffered (preferably pH greater than 8), if necessary, and the liquid diluent first rendered isotonic. The aqueous solutions are suitable for intravenous injection purposes. The preparation of all these solutions under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • The following non-limitative examples further describe and enable a person ordinarily skilled in the art to make and use the invention.
  • Preparation of Compounds EXAMPLE 1 Synthesis of trimebutine nitroargininate (I)
  • Figure US20110144188A1-20110616-C00007
  • To a mixture of H-Arg(NO2)—OH (0.1 mole) and trimebutine (0.1 mole), water (200 mL) and ethyl alcohol (20 mL) have been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • 1H-NMR (400 MHz, DMSO-d6): δ 0.60 (t, 3H), 1.45-1.75 (m, 4H), 1.80-1.90 (m, 2H), 2.25 (s, 6H), 2.90-3.40 (m, 2H), 3.75 (s, 9H), 3.95 (m, 1H), 4.64 (dd, 2H), 7.15 (s, 2H), 7.22 (t, 1H), 7.35 (t, 2H), 7.46 (d, 2H).
  • 13C-NMR (400 MHz, DMSO-d6): δ 9.07, 22.8, 26.4, 28.9, 29.1, 47.9, 56.4, 60.8, 64.4, 65.8, 107.3, 125.2, 127.4, 128.0, 128.5, 141.7, 142.5, 153.4, 158.3, 165.9, 170.2.
  • mp 183° C. (dec).
  • EXAMPLE 2 Synthesis of trimebutine cysteinyl-nitroargininate (II)
  • Figure US20110144188A1-20110616-C00008
  • Synthesis of 2-(2-amino-3-mercapto-propionylamino)-5-nitroguanidino-pentanoic acid (b)
  • To a solution of Boc-Cys(Trt)-OH (3.0 mmol) in 50 mL of dimethylformamide, hydroxybenzotriazole (3.3 mmol) and DCC (3.3 mmol) were added with stirring at 0° C. for 1 h. To the reaction mixture, H-Arg(NO2)-OtBu (3.0 mmol) was added and stirred mechanically for 3 h at 0° C. and 24 h at room temperature. After filtration, the filtrate was evaporated under reduced pressure to remove the solvent. The oily residue thus obtained was dissolved in ethyl acetate; the organic layer was washed with brine, dried on anhydrous MgSO4, filtered and the solvent evaporated. The crude intermediate a was treated with a solution of trifluoroacetic acid in dichloromethane (40% TFA in DCM). After 1 h the solvent was removed to obtain H-Cys-Arg(NO2)—OH.TFA as a crude residue which was precipitated with diethyl ether; the obtained solid was dissolved in water and 1N NaOH was slowly added to obtain 2-(2-amino-3-mercapto-propionylamino)-5-nitroguanidino-pentanoic acid (b) as a white solid which was recovered by filtration.
  • Synthesis of trimebutine cysteinyl-nitroargininate (II)
  • To a mixture of 2-(2-amino-3-mercapto-propionylamino)-5-nitroguanidino-pentanoic acid (b; 0.1 mole) and trimebutine (0.1 mole), water (200 mL) and ethyl alcohol (20 mL) have been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • EXAMPLE 3 Synthesis of trimebutine thiocarbamoylbenzoate (III) Preparation of 3,4,5-trimethoxybenzoic acid 2-(dimethylamino)-2-phenylbutyl ester 4-thiocarbamoyl benzoate (Trimebutine thiocarbamoylbenzoate)
  • Figure US20110144188A1-20110616-C00009
  • To a mixture of 4-(thiocarbamoyl)benzoic acid (0.1 mol) and trimebutine (0.1 mol), water (200 mL) and ethyl alcohol (20 mL) have been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • 1H-NMR (400 MHz, DMSO-d6): δ 0.60 (t, 3H), 1.45-1.75 (m, 4H), 1.80-1.90 (m, 2H), 2.28 (s, 6H), 2.90-3.40 (m, 2H), 3.69 (s, 9H), 3.95 (m, 1H), 4.73 (dd, 2H), 7.01 (s, 2H), 7.22 (t, 1H), 7.35 (t, 2H), 7.46 (d, 2H) 7.93 (dd, 4H), 9.65 (bs, 1H, NH), 10.05 (bs, 1H, NH).
  • 13C-NMR (400 MHz, DMSO-d6): δ 9.07, 28.9, 56.5, 60.8, 64.5, 65.7, 107.1, 125.3, 127.4, 128.1, 128.6, 129.5, 129.7, 132.3, 141.8, 142.5, 148.5, 153.4, 154.8, 165.9, 169.4, 172.5, 188.6.
  • mp 66-68° C. (dec).
  • Synthesis of 4-(thiocarbamoyl)benzoic acid
  • The compound was synthesized according to a procedure already reported in literature (Fairfull, E. S., Lowe J. L., Peak D. A. J. Chem. Soc. 1952, 742), incorporated herein by reference.
  • Figure US20110144188A1-20110616-C00010
  • 4-(Thiocarbamoyl)benzoic acid (2)
  • 3 g of 4-cyanobenzoic acid 1 (20.4 mmol) were dissolved in 40 mL of pyridine and 2.1 mL of triethylamine (20.4 mmol) were added. Dry hydrogen sulphide was passed through the solution in a steady steam for 4 h. The mixture was then poured into water and the solid collected by filtration. Recrystallization from petroleum ether furnished 2.51 g of the pure compound 2 (68%yield).
  • MS (ESI), m/e 182.2 (M+).
  • 1H NMR (DMSO-d6): δ 7.92 (dd, 4H), 9.68 (s, 1H, NH), 10.12 (s, 1H, NH), 13.25 (s, 1H, OH).
  • 13C NMR (DMSO-d6): δ 127.3, 129. 6, 132.0, 148.5, 169.4, 188.6
  • m.p. 296-298° C. (dec.)
  • EXAMPLE 4 Synthesis of trimebutine ADT-nitroargininate (IV)
  • Figure US20110144188A1-20110616-C00011
  • Synthesis of carbonic acid 4-nitro-phenyl ester 4-(5-thioxo-5H-[1,2]dithiol-3-yl)-phenyl ester (a)
  • To a stirred suspension of ADT-OH (1.04 mmol) in CH2Cl2 (10 ml) was added 4-dimethylaminopyridine. (DMAP, 1.16 mmol) and 4-nitrophenyl chloroformate (1.15 mmol). The reaction mixture was stirred for 10 hr at room temperature. Thin layer chromatography indicated that the formation of the desired product completed. The solvent was removed and the residue was treated with diethyl ether; product a was recovered by filtration and used without further purification (yield 81%).
  • Synthesis of 5-nitroguanidino-2-[4-(5-thioxo-5H-[1,2]dithiol-3-yl)-phenoxycarbonyl-amino]-pentanoic acid (b)
  • To the solution of a (1.04 mmol) in 50 mL of CH2Cl2 were added 4-dimethylaminopyridine (1.16 mmol) and H-Arg(NO2)-OtBu (1.02 mmol) and the solution was stirred for 20 hr at room temperature. Then, the reaction mixture was diluted with CH2Cl2, washed with sat. NaHCO3 and sat. NaCl, and dried over MgSO4. The crude intermediate was treated with a solution of trifluoroacetic acid in dichloromethane (40% TFA in DCM). After 1 h the solvent was removed to obtain product b as a crude residue which was precipitated with diethyl ether; the obtained solid was dissolved in water and 1N NaOH was slowly added to obtain 5-nitroguanidino-2-[4-(5-thioxo-5H-[1,2]dithiol-3-yl)-phenoxycarbonyl-amino]-pentanoic acid (b) as a white solid which was recovered by filtration.
  • Synthesis of trimebutine ADT-nitroargininate (IV)
  • To a mixture of 5-nitroguanidino-2-[4-(5-thioxo-5H-[1,2]dithiol-3-yl)-phenoxycarbonyl-amino]-pentanoic acid (b; 0.1 mol) and trimebutine (0.1 mol), water (200 mL) and ethyl alcohol (20 mL) have been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • EXAMPLE 5 Synthesis of trimebutine p-hydroxythiobenzamide-nitroargininate (V)
  • Figure US20110144188A1-20110616-C00012
  • Synthesis of carbonic acid 4-nitro-phenyl ester 4-thiocarbamoyl-phenyl ester (a)
  • To a stirred suspension of p-hydroxythiobenzamide (1.04 mmol) in CH2Cl2 (10 ml) was added 4-dimethylaminopyridine (DMAP, 1.16 mmol) and 4-nitrophenyl chloroformate (1.15 mmol). The reaction mixture was stirred for 10 hr at room temperature. Thin layer chromatography indicated that the formation of the desired product completed. The solvent was removed and the residue was treated with diethyl ether; product a was recovered by filtration and used without further purification (yield 81%).
  • Synthesis of 5-nitroguanidino-2-(4-thiocarbamoyl-phenoxycarbonylamino)-pentanoic acid (b)
  • To the solution of a (1.04 mmol) in 50 mL of CH2Cl2 were added 4-dimethylaminopyridine (1.16 mmol) and H-Arg(NO2)-OtBu (1.02 mmol) and the solution was stirred for 20 hr at room temperature. Then, the reaction mixture was diluted with CH2Cl2, washed with sat. NaHCO3 and sat. NaCl, and dried over MgSO4. The crude intermediate was treated with a solution of trifluoroacetic acid in dichloromethane (40% TFA in DCM). After 1 h the solvent was removed to obtain product b as a crude residue which was precipitated with diethyl ether; the obtained solid was dissolved in water and 1N NaOH was slowly added to obtain 5-nitroguanidino-2-(4-thiocarbamoyl-phenoxycarbonylamino)-pentanoic acid (b) as a white solid which was recovered by filtration.
  • Synthesis of trimebutine p-hydroxythiobenzamide-nitroargininate (V)
  • To a mixture of 5-nitroguanidino-2-(4-thiocarbamoyl-phenoxycarbonylamino)-pentanoic acid (b; 0.1 mole) and trimebutine (0.1 mole), water (200 mL) and ethyl alcohol (20 mL) have been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • EXAMPLE 6 Synthesis of N-desmethyltrimebutine nitroargininate (VI)
  • Figure US20110144188A1-20110616-C00013
  • To a mixture of H-Arg(NO2)—OH (0.1 mol) and N-desmethyltrimebutine (0.1 mol), water (200 mL) and ethyl alcohol (20 mL) have been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • 1H-NMR (400 MHz, DMSO-d6): δ 0.72 (t, 3H), 1.45-1.75 (m, 4H), 1.80-1.90 (m, 2H), 2.07 (s, 3H), 2.90-3.40 (m, 2H), 3.75 (s, 9H), 3.95 (m, 1H), 4.64 (dd, 2H), 7.07 (s, 2H), 7.22 (t, 1H), 7.35 (t, 2H), 7.51 (d, 2H).
  • 13C-NMR (400 MHz, DMSO-d6): δ 9.07, 22.8, 26.4, 28.9, 29.1, 47.9, 56.4, 60.8, 64.4, 65.8, 107.3, 125.2, 127.4, 128.0, 128.5, 141.7, 142.5, 153.4, 158.3, 165.9, 170.2.
  • m.p. 78-80° C. (dec)
  • Synthesis of N-desmethyltrimebutine
  • The compound was synthesized following with slight modifications the procedure reported in literature (Martin, A., Figadere B., Saivin S., Houin G., Chomard J. M., Cahiez G. Arzneim.-Forsch./Drug Res. 2000 (50), 544).
  • Figure US20110144188A1-20110616-C00014
    Figure US20110144188A1-20110616-C00015
  • Phenylglycin ethylester hydrochloride (2)
  • Figure US20110144188A1-20110616-C00016
  • 22 mL of SOCl2 were added dropwise to a solution of 30 g of phenylglycin 1 (198.5 mmol) in 200 mL of anhydrous ethanol. A gentle reflux occurred spontaneously and it was maintained for 3 h. The reaction was allowed to cool to room temperature and was stirred overnight. The solvent was removed under vacuum to afford 41.8 g of 2 as a white powder (98% yield).
  • MS (ESI), m/e 179.8 (M+).
  • Ethyl N-(phenylmethylene)glycinate (3)
  • Figure US20110144188A1-20110616-C00017
  • To a mixture of 10.6 g of ethyl phenylglycinate hydrochloride 2 (49.3 mmol), 100 mL of dichloromethane, 5 ml of benzaldehyde (49.2 mmol) and 30 g of magnesium sulfate (249.2 mmol) were added at room temperature, under a nitrogen atmosphere, 21.25 mL of triethylamine (152.46 mmol). After stirring for 17 h the heterogeneous reaction mixture was filtered through celite and the solid was washed with 100 mL of dichloromethane. The solvents were evaporated under reduced pressure and the viscous oil thus obtained was stirred with 80 mL of diethyl ether and 80 mL of water until dissolution. After decantation, the organic layer was dried over sodium sulphate and the solvent was evaporated under vacuum. The immine ester 3 (12.4 g, 94% yield) was obtained as a yellow pale oil.
  • MS (ESI), m/e 268.3 (M+).
  • Ethyl 2-phenyl-2-(N-phenylmethylene)-butanoate (4)
  • Figure US20110144188A1-20110616-C00018
  • Under a nitrogen atmosphere, a solution of 12.39 g (46.4 mmol) of 3 in 64 mL of anhydrous THF was added dropwise, under stirring, to a mixture of 2.04 of sodium hydride (60% oil dispersion, 85 mmol), 192 mg of CuCl2 and 128 mL of anhydrous THF. After 9 h at room temperature, 4.66 mL (57.7 mmol) of ethyl iodide were quickly added to the reaction mixture. Stirring was continued for 18 h and 0.86 mL of anhydrous ethanol were added cautiously to the reaction mixture. During the stirring the reaction colour changed from yellow to red to orange and then to green. After concentration of the reaction mixture under vacuum, 95.8 mL of diethyl ether and 160 mL water were added and the resulting mixture was stirred for 10 minutes, then filtered through celite. After decantation the organic layer was washed three times with water and dried over sodium sulphate, then the diethyl ether was evaporated under reduced pressure to furnish 5.82 g of 4 (43% yield). The crude product, obtained as a yellow-orange oil, was pure enough to be used without further purification.
  • MS (ESI), m/e 296.1 (M+).
  • Ethyl 2-amino-2-phenylbutanoate hydrochloride (5)
  • Figure US20110144188A1-20110616-C00019
  • 5.8 g of 4 (19.73 mmol), 17.6 mL of THF, 35.17 mL of diethyl ether, 44 mL of water and 2.64 mL of concentrated HCl were stirred at room temperature for 24 h. Solvents were removed from reaction mixture under vacuum and the resulting aqueous solution was washed twice with diethyl ether. Water was then evaporated under reduced pressure to give 3.8 g of 5 as an orange oil (79% yield).
  • Ethyl 2-amino-2-phenylbutanoate (6)
  • Figure US20110144188A1-20110616-C00020
  • The crude product 5 was dissolved in 22 mL of anhydrous THF and 2.64 mL of triethyl amine were added. After stirring for 30 minutes, the mixture was filtered through celite and the solid washed with 100 mL of anhydrous diethyl ether. Concentration of the solvents using a rotary evaporator afforded 2.49 g of 6 as a blue oil (77% yield).
  • MS (ESI), m/e 208 (M+).
  • Ethyl 2-Formylamino-2-phenylbutanoate (7)
  • Figure US20110144188A1-20110616-C00021
  • 12.72 mL of formic acid then 25.38 mL of acetic anhydride were added dropwise, under stirring, to 2.49 g (12.03 mmol) of amine 6. After 15 h at room temperature, solvents were removed under reduced pressure to give 3.09 g of 7 as a green viscous oil (100% yield).
  • Rf=0.85 (CHCl3/MeOH 9.5:0.5)
  • MS (ESI), m/e 236 (M+).
  • 2-Methylamino-2-phenylbutanol (8)
  • Figure US20110144188A1-20110616-C00022
  • To a suspension of 900 mg (23.71 mmol) of lithium aluminium hydride (LiAlH4) in 30 mL of anhydrous THF were added dropwise a solution of 3.09 g (13.15 mmol) of 7, previously prepared, in 18 mL of anhydrous THF. After 4 h under reflux, the reaction mixture was allowed to cool to room temperature and an additional amount of lithium aluminium hydride (900 mg) was introduced. Reflux was maintained for 4 h then an aqueous saturated solution of magnesium sulfate was slowly added at −10° C. under vigorous stirring until formation of a precipitate. The solid was filtered and washed several times with THF. The organic solvent was evaporated under reduced pressure until obtaining of an aqueous solution. The solution was added with 100 mL of diethyl ether and the organic layer was dried over sodium sulfate. Evaporation of the solvent afforded 1.84 g of 8 as a yellow oil. (78% yield).
  • MS (ESI), m/e 180.1 (M+).
  • (2-Methylamino-2-phenylbutyl)3,4,5-trimethoxybenzoate (N-desmethyltrimebutine (9)
  • Figure US20110144188A1-20110616-C00023
  • To a solution of 1.84 g of 8 (10.3 mmol) in 28 mL of anhydrous THF were added dropwise, at −78° C., 4.11 mL of n-BuLi (2.5 mol/L in hexanes). After 15 minutes, a solution of 2.31 g (10.01 mmol) of 3,4,5-trimethoxybenzoyl chloride in 15.8 mL of anhydrous THF was added. The reaction mixture was then allowed to warm to −30° C. (ca 1 h) and 19 mL of acetic acid were cautiously added. Solvents were removed under vacuum and 55 mL of diethyl ether and 55 mL of water were added to the previously obtained oil. After stirring until complete dissolution, the resulting mixture was then decanted and the aqueous layer alcalinized completely with solid Na2CO3 and re-extracted with diethyl ether. The organic layers were washed twice with a saturated aqueous solution of sodium carbonate and with brine and then dried over sodium sulphate. Diethyl ether was evaporated under vacuum and the ester 9 was purified by chromatography on silica gel column (ethyl acetate/n-hexane, 7:3) to give 1.4 g of pure N-desmethyltrimebutine 9 (49% yield) as a yellow pale oil. MS (ESI), m/e 374.1 (M+).
  • 1H NMR (CDCl3): δ 0.72 (t, 3H), 1.7 (s, 1H, NH), 1.75-1.9 (m, 2H), 2.6 (s, 3H), 3.76 (s, 6H), 3.80 (s, 3H), 4.50 (dd, 2H), 7.07-7.44 (m, 7H).
  • 13C NMR (DMSO-d6): δ 7.4, 28.5, 28.6, 55.9, 60.1, 61.1, 66.3, 100.5, 126.5, 127.0, 129.3, 128.0, 142.0, 142.5, 152.7, 165.6.
  • EXAMPLE 7 Synthesis of N-desmethyltrimebutine cysteinyl-nitroargininate (VII)
  • Figure US20110144188A1-20110616-C00024
  • Synthesis of 2-(2-amino-3-mercapto-propionylamino)-5-nitroguanidino-pentanoic acid (b)
  • To a solution of Boc-Cys(Trt)-OH (3.0 mmol) in 50 mL of dimethylformamide, hydroxybenzotriazole (3.3 mmol) and DCC (3.3 mmol) were added with stirring at 0° C. for 1 h. To the reaction mixture, H-Arg(NO2)-OtBu (3.0 mmol) was added and stirred mechanically for 3 h at 0° C. and 24 h at room temperature. After filtration, the filtrate was evaporated under reduced pressure to remove the solvent. The oily residue thus obtained was dissolved in ethyl acetate; the organic layer was washed with brine, dried on anhydrous MgSO4, filtered and the solvent evaporated. The crude intermediate a was treated with a solution of trifluoroacetic acid in dichloromethane (40% TFA in DCM). After 1 h the solvent was removed to obtain H-Cys-Arg(NO2)-OH.TFA as a crude residue which was precipitated with diethyl ether; the obtained solid was dissolved in water and 1N NaOH was slowly added to obtain 2-(2-amino-3-mercapto-propionylamino)-5-nitroguanidino-pentanoic acid (b) as a white solid which was recovered by filtration.
  • Synthesis of N-desmethyltrimebutine cysteinyl-nitroargininate (VII)
  • To a mixture of 2-(2-amino-3-mercapto-propionylamino)-5-nitroguanidino-pentanoic acid (b; 0.1 mole) and N-desmethyltrimebutine (0.1 mole), water (200 mL) and ethyl alcohol (20 mL) have been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • EXAMPLE 8 Preparation of 3,4,5-trimethoxybenzoic acid 2-(methylamino)-2-phenylbutyl ester 4-thiocarbamoyl benzoate (N-desmethyltrimebutine thiocarbamoylbenzoate (VIII)
  • Figure US20110144188A1-20110616-C00025
  • A mixture of 4-(thiocarbamoyl)benzoic acid (0.1 mol) and desmethyltrimebutine (0.1 mol) was dissolved in ethyl alcohol (20 mL) and acetonitrile (20 mL), then water (200 mL) has been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • 1H-NMR (400 MHz, DMSO-d6): δ 0.72 (t, 3H), 1.70-1.80 (m, 4H), 1.80-1.90 (m, 2H), 2.08 (s, 3H), 2.90-3.40 (m, 2H), 3.69 (s, 9H), 3.95 (m, 1H), 4.41 (dd, 2H), 7.07 (s, 2H), 7.22 (t, 1H), 7.33 (t, 2H), 7.52 (d, 2H) 7.93 (dd, 4H), 9.63 (bs, 1H, NH), 10.02 (bs, 1H, NH).
  • 13C-NMR (400 MHz, DMSO-d6): δ 9.07, 28.7, 56.5, 60.5, 64.6, 65.8, 107.2, 125.5, 127.2, 128.2, 128.6, 129.5, 129.4, 132.5, 141.9, 142.4, 148.5, 153.5, 154.7, 165.7, 169.4, 172.5, 188.6.
  • mp 65-67° C. (dec.)
  • EXAMPLE 9 Synthesis of N-desmethyltrimebutine ADT-nitroargininate (IX)
  • Figure US20110144188A1-20110616-C00026
  • Synthesis of carbonic acid 4-nitro-phenyl ester 4-(5-thioxo-5H-[1,2]dithiol-3-yl)-phenyl ester (a)
  • To a stirred suspension of ADT-OH (1.04 mmol) in CH2Cl2 (10 ml) was added 4-dimethylaminopyridine (DMAP, 1.16 mmol) and 4-nitrophenyl chloroformate (1.15 mmol). The reaction mixture was stirred for 10 hr at room temperature. Thin layer chromatography indicated that the formation of the desired product completed. The solvent was removed and the residue was treated with diethyl ether; product a was recovered by filtration and used without further purification (yield 81%).
  • Synthesis of 5-nitroguanidino-2-[4-(5-thioxo-5H-[1,2]dithiol-3-yl)-phenoxycarbonyl-amino]-pentanoic acid (b)
  • To the solution of a (1.04 mmol) in 50 mL of CH2Cl2 were added 4-dimethylaminopyridine (1.16 mmol) and H-Arg(NO2)-OtBu (1.02 mmol) and the solution was stirred for 20 hr at room temperature. Then, the reaction mixture was diluted with CH2Cl2, washed with sat. NaHCO3 and sat. NaCl, and dried over MgSO4. The crude intermediate was treated with a solution of trifluoroacetic acid in dichloromethane (40% TFA in DCM). After 1 h the solvent was removed to obtain product b as a crude residue which was precipitated with diethyl ether; the obtained solid was dissolved in water and 1N NaOH was slowly added to obtain 5-nitroguanidino-2-[4-(5-thioxo-5H-[1,2]dithiol-3-yl)-phenoxycarbonyl-amino]-pentanoic acid (b) as a white solid which was recovered by filtration.
  • Synthesis of N-desmethyltrimebutine ADT-nitroargininate (IX)
  • To a mixture of 5-nitroguanidino-2-[4-(5-thioxo-5H-[1,2]dithiol-3-yl)-phenoxycarbonyl-amino]-pentanoic acid (b; 0.1 mol) and N-desmethyltrimebutine (0.1 mol), water (200 mL) and ethyl alcohol (20 mL) have been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • EXAMPLE 10 Synthesis of N-desmethyltrimebutine p-hydroxythiobenzamide nitro-argininate (X)
  • Figure US20110144188A1-20110616-C00027
  • Synthesis of carbonic acid 4-nitro-phenyl ester 4-thiocarbamoyl-phenyl ester (a)
  • To a stirred suspension of p-hydroxythiobenzamide (1.04 mmol) in CH2Cl2 (10 ml) was added 4-dimethylaminopyridine (DMAP, 1.16 mmol) and 4-nitrophenyl chloroformate (1.15 mmol). The reaction mixture was stirred for 10 hr at room temperature. Thin layer chromatography indicated that the formation of the desired product completed. The solvent was removed and the residue was treated with diethyl ether; product a was recovered by filtration and used without further purification (yield 81%).
  • Synthesis of 5-nitroguanidino-2-(4-thiocarbamoyl-phenoxycarbonylamino)-pentanoic acid (b)
  • To the solution of a (1.04 mmol) in 50 mL of CH2Cl2 were added 4-dimethylaminopyridine (1.16 mmol) and H-Arg(NO2)-OtBu (1.02 mmol) and the solution was stirred for 20 hr at room temperature. Then, the reaction mixture was diluted with CH2Cl2, washed with sat. NaHCO3 and sat. NaCl, and dried over MgSO4. The crude intermediate was treated with a solution of trifluoroacetic acid in dichloromethane (40% TFA in DCM). After 1 h the solvent was removed to obtain product b as a crude residue which was precipitated with diethyl ether; the obtained solid was dissolved in water and 1N NaOH was slowly added to obtain 5-nitroguanidino-2-(4-thiocarbamoyl-phenoxycarbonylamino)-pentanoic acid (b) as a white solid which was recovered by filtration.
  • Synthesis of N-desmethyltrimebutine p-hydroxythiobenzamide-nitroargininate (X)
  • To a mixture of 5-nitroguanidino-2-(4-thiocarbamoyl-phenoxycarbonylamino)-pentanoic acid (b; 0.1 mole) and N-desmethyltrimebutine (0.1 mole), water (200 mL) and ethyl alcohol (20 mL) have been added and the resulting suspension has been stirred at room temperature until clear. Then the solution has been frozen and lyophilized furnishing the desired salt (quantitative yield).
  • Testing of Compounds EXAMPLE 11 Comparison of the Effects of Salt I, Trimebutine Nitroargininate, Versus Trimebutine Alone and Nitroarginine Alone in a Rat Model of Visceral Pain Perception
  • A rat model of visceral pain perception, a pre-clinical model of irritable bowel syndrome, was used in the following example. Rats (male, Wistar, 200-250 g, obtained from Charles River, Monza, Italy), were housed in plastic cages and maintained under controlled conditions with 12-hours light/dark cycle with lights on at 7.00 AM. Tap water and standard laboratory chow were freely available. Before experiments, rats were individually trained by spending 2-3 hours per day in a plexiglass cage for 2-3 days. It allowed them to adjust to a movement-restriction environment. Food was withheld for 12 hours before colorectal distension (CRD) recording were performed. Experiments were performed in awake rats and were conducted in a blind manner in that the observer was not aware of the identity of drug administered to each animal.
  • On the testing day, rats were sedated with ether inhalation and a 2 cm long latex balloon was inserted intrarectally 2 cm from the anal verge and fixed at the base of the tail. The balloon was connected via a double-barreled cannula to a pressure transducer to continuously monitoring the rectal pressure by a computer (PowerLab PC, A.D. Instruments, Milford, Mass., USA) and to a syringe for inflation/deflation of the balloon. The rats were then housed in a small cage (20×8×8 cm) on an elevated Plexiglas™ platform and allowed to wake up and adapt for 1 hour. After recovery from sedation, animals underwent the CRD procedure and behavioral responses were tested. The night before the experiments, the balloons were inflated and left overnight so the latex stretched and the balloons became compliant.
  • CRD of 20 seconds, performed every 5 minutes, was applied in increment of 0.4 ml starting from 0.4 ml up to 1.6 ml water. To achieve an accurate measurement of the colonic parameters and perception, the distensions were repeated twice for each intensity and data for each animal were averaged for analysis. Each animal underwent a double set of CRD. Twenty minutes after the first sequence of CRD (0.4 mL-1.6 ml water), drugs were administered intraperitoneally (i.p.) and a second set of CRD was performed. Behavioral responses during the first and the second set of CRD were assessed and compared.
  • Behavioral response to CRD was assessed by measuring the abdominal withdrawal reflex (AWR) using a semi-quantitative score (1). The AWR is an involuntary motor reflex similar to the visceromotor reflex, but it has the great advantage that, in contrast to the latter, it does not require abdominal surgery to implant recording electrodes and wires in the abdominal muscle wall which may cause additional sensitization (see Ness, T. J. and Gebhart, G. F. (1990) Pain 41:167-234, incorporated herein by reference).
  • Measurement of the AWR consisted of visual observation of the animal response to graded CRD by blinded observer and assignment of an AWR score according with the behavioral scale as previously described in Al-Chaer, E. D. et al. (2000) Gastroenterology 19: 1276-85, incorporated herein by reference, in which grade 0 corresponds to no behavioral response to CRD, grade 1 corresponds to brief head movement at the onset of the stimulus followed by immobility, grade 2 corresponds to a mild contraction of abdominal muscles although the rats does not lift the abdomen off the platform, grade 3 corresponds to a strong contraction of the abdominal muscles with the lifting of the abdomen off the platform, and grade 4 corresponds to a severe contraction of the abdominal muscle manifested by body arching and the lifting of the abdomen and of the pelvic structures and scrotum.
  • The effects of trimebutine maleate, nitroarginine and trimebutine nitroargininate on colonic compliance and sensitivity were determined using a total of 15 fasting rats. To investigate whether the administration of trimebutine maleate, nitroarginine and trimebutine nitroargininate could reduce pain induced by CRD, after the first sequence of CRD (vehicle-treated), 5 rats were treated with trimebutine maleate at a dose of 10 mg/kg i.p., nitroarginine at a dose of 6 mg/kg or trimebutine nitroargininate at the dose of 16 mg/kg i.p., after which a second set of CRD was repeated. Results from these experiments are shown in FIGS. 1( a), 2(a) and 3(a).
  • To determine the effect of trimebutine maleate, nitroarginine and trimebutine nitroargininate on colonic smooth muscle, the compliance of the colo-rectum during CRD was obtained from intracolo-rectal volume and pressure and expressed as mL/mmHg. These results are shown in FIGS. 1( b), 2(b) and 3(b).
  • To determine the role of NO in the visceral analgesic effects of trimebutine nitroargininate, experiments were performed in which rats were pretreated 10 min before administration of trimebutine nitroargininate (16 mg/kg i.p.) or vehicle with methylene blue (1 mg/kg i.p.), L-NAME (25 mg/kg i.v.) or vehicle. The results are shown in FIGS. 4( a) and 4(b).
  • All data are presented as the mean±SEM, with sample sizes of 5 rats/group; statistical comparison of data was performed by the Student's paired t-test. An associated probability (p value) of less that 5% was considered significant, as indicated by an asterisk.
  • FIGS. 1( a), 2(a) and 3(a) show that trimebutine nitroargininate is more effective than either trimebutine maleate or nitroarginine in reducing visceral pain in response to colorectal distension. However, none of the compounds were particularly effective in reducing intrarectal pressure, as shown in FIGS. 1( b), 2(b) and 3(b). FIGS. 4( a) and 4(b) show that the visceral analgesic effects of trimebutine nitroargininate are largely abolished by pretreatment with an inhibitor of nitric oxide synthase (L-NAME) or with an inhibitor of soluble guanylate cyclase (methylene blue). These results suggest that release of nitric oxide from trimebutine nitroargininate, and stimulation of soluble guanylate cyclase, contribute significantly to the visceral analgesic effects of this compound.
  • Thus, trimebutine nitroargininate is useful in treating abdominal pain associated with various inflammatory conditions of the alimentary tract, as well as functional gastrointestinal disorders such as irritable bowel syndrome, dyspepsia, etc., that are characterized by increased visceral nociception (with or without accompanying inflammation).
  • EXAMPLE 12 Comparison of the Effects of Salt III, Trimebutine Thiocarbamoylbenzoate, Versus Trimebutine Alone and Thiocarbamoylbenzoate Alone, in a Rat Model of Visceral Pain Perception
  • Experiments were carried out as described in Example 11, except that groups of 5 rats each were treated with vehicle, trimebutine maleate (10 mg/kg), or with equimolar doses of trimebutine thiocarbamoylbenzoate (salt III) or thiocarbamolybenzoate alone.
  • FIGS. 5( a) and 5(b) show that trimebutine thiocarbamoylbenzoate is more effective than either trimebutine maleate or thiocarbamoylbenzoate in reducing visceral pain in response to colorectal distension.
  • Thus, trimebutine thiocarbamoylbenzoate is useful in treating abdominal pain associated with various inflammatory conditions of the alimentary tract, as well as functional gastrointestinal disorders such as irritable bowel syndrome, dyspepsia, etc., that are characterized by increased visceral nociception (with or without accompanying inflammation).
  • EXAMPLE 13 Generation of H2S by 4-(thiocarbamoyl)benzoic acid and 5-(4-Amino-phenyl)-[1,2]dithiole-3-thione
  • Two compounds were tested, 5-(4-Amino-phenyl)-[1,2]dithiole-3-thione (ADT-OH) and 4-(thiocarbamoyl)benzoic acid (TBZ) for H2S generation under three different conditions for H2S generation. In particular, concentrations of H2S generated within 1 hour from 1 mM concentrations ADT-OH and TBZ were measured. H2S release was tested under three conditions: (i) when the compound was in buffer, (ii) when the compound was in a liver homogenate, and (iii) when the compound was in the liver homogenate together with an inhibitor of H2S synthesis, which blocks the activity of the enzyme an inhibitor of cystathionine-γ-lyase (PAG=DL-propargylglycine; 2 mM). Results are shown in FIG. 6. Asterisk (*) indicates a significant (p<0.05) increase versus corresponding vehicle-treated group. The alpha (α) represents a significant decrease in H2S synthesis as a result of incubation in the presence of PAG.

Claims (21)

1. A compound of the general formula:

A+·X
where:
A is
Figure US20110144188A1-20110616-C00028
and their corresponding stereoisomers;
and X is a NO-releasing, an H2S-releasing moiety, or a combined NO- and H2S-releasing moiety.
2. The compound of claim 1, wherein X is selected from the group consisting of:
Figure US20110144188A1-20110616-C00029
3. The compound according to claim 2 where the compound is trimebutine nitroargininate.
4. The compound according to claim 2 where the compound is trimebutine cysteinyl-nitroargininate.
5. The compound according to claim 2 where the compound is trimebutine thiocarbamoyl benzoate.
6. The compound according to claim 2 where the compound is trimebutine 5-phenyl-1,2-dithione-3-thione-nitroargininate.
7. The compound according to claim 2 where the compound is trimebutine p-hydroxythiobenzamide-nitroargininate.
8. The compound according to claim 2 where the compound is N-desmethyltrimebutine nitroargininate.
9. The compound according to claim 2 where the compound is N-desmethyltrimebutine cysteinyl-nitroargininate.
10. The compound according to claim 2 wherein the compound is N-desmethyltrimebutine thiocarbamoylbenzoate.
11. The compound according to claim 2 wherein the compound is N-desmethyltrimebutine 5-phenyl-1,2-dithione-3-thione-nitroargininate
12. The compound according to claim 2 wherein the compound is N-desmethyltrimebutine p-hydroxythiobenzamide-nitroargininate.
13. A pharmaceutical composition comprising a compound as claimed in claim 2 and a pharmaceutically acceptable excipient or carrier.
14. A method for treating visceral pain in a subject in need of such treatment comprising administering to the subject a visceral pain relieving amount of a compound according to claim 2.
15. A method as claimed in claim 14, wherein the visceral pain is abdominal pain.
16. A method as claimed in claim 15, wherein the abdominal pain is due to intestinal diseases such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), diabetic gastroparesis, and dyspepsia.
17. A compound having the general formula nitroarginine-R, wherein R is an H2S-releasing moiety, for the preparation of salts of trimebutine and N-monodesmethyl trimebutine.
18. A compound as claimed in claim 17, wherein R is selected from the group consisting of 5-p-hydroxyphenyl-1,2-dithione-3-thione, cysteine, and 4-(thiocarbamoyl)benzoic acid.
19. (canceled)
20. (canceled)
21. (canceled)
US12/303,082 2006-06-06 2007-06-06 Salts of trimebutine and n-desmethyl trimebutine Abandoned US20110144188A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/303,082 US20110144188A1 (en) 2006-06-06 2007-06-06 Salts of trimebutine and n-desmethyl trimebutine

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US80406706P 2006-06-06 2006-06-06
US12/303,082 US20110144188A1 (en) 2006-06-06 2007-06-06 Salts of trimebutine and n-desmethyl trimebutine
PCT/CA2007/001008 WO2007140611A1 (en) 2006-06-06 2007-06-06 Salts of trimebutine and n-desmethyl trimebutine

Publications (1)

Publication Number Publication Date
US20110144188A1 true US20110144188A1 (en) 2011-06-16

Family

ID=38801016

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/759,154 Expired - Fee Related US7666907B2 (en) 2005-05-27 2007-06-06 Salts of trimebutine and N-desmethyl trimebutine
US12/303,082 Abandoned US20110144188A1 (en) 2006-06-06 2007-06-06 Salts of trimebutine and n-desmethyl trimebutine

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/759,154 Expired - Fee Related US7666907B2 (en) 2005-05-27 2007-06-06 Salts of trimebutine and N-desmethyl trimebutine

Country Status (10)

Country Link
US (2) US7666907B2 (en)
EP (1) EP2024323B1 (en)
JP (1) JP5421098B2 (en)
KR (1) KR20090021346A (en)
CN (1) CN101489986A (en)
AU (1) AU2007257174B2 (en)
BR (1) BRPI0712417A2 (en)
CA (1) CA2653869A1 (en)
RU (1) RU2008152751A (en)
WO (1) WO2007140611A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7741359B2 (en) * 2005-05-27 2010-06-22 Antibe Therapeutics Inc. Hydrogen sulfide derivatives of non-steroidal anti-inflammatory drugs
US7932286B2 (en) * 2007-07-31 2011-04-26 Martin Francois Method for treating colonic viscerosensitivity and spasticity
FR2927076B1 (en) * 2008-01-31 2010-03-26 Oroxcell 2-AMINO-2-PHENYL-ALKANOL DERIVATIVES, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US9688607B2 (en) 2011-08-15 2017-06-27 Research Foundation Of The City University Of New York No- and H2S-releasing compounds
CN104254517A (en) * 2012-03-12 2014-12-31 吉卡尔制药公司 Novel sulfonate-based trimebutine salts
EP3197443A4 (en) * 2014-09-25 2018-03-14 The Hospital for Sick Children Compositions and methods for enhancing hair growth, promoting skin regeneration, and wound healing
RU2752086C1 (en) * 2020-04-27 2021-07-22 Общество С Ограниченной Ответственностью "Валента-Интеллект" New [2-(dimethylamino)-2-phenylbutyl]-3,4,5-trimetoxybenzoate 4-methyl-2h-chromen-2-on-7-ilsulphate compound and application thereof
CN114916547A (en) * 2021-12-06 2022-08-19 南京天秾生物技术有限公司 Application of 2, 6-diamino-3-methylhexanoic acid as plant immune inducer

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4301163A (en) * 1979-07-11 1981-11-17 Societe Industrielle De Produits De Synthese Amino-ether oxides and use thereof in therapy
US4412992A (en) * 1980-07-21 1983-11-01 Biorex Laboratories Limited 2-Hydroxy-5-phenylazobenzoic acid derivatives and method of treating ulcerative colitis therewith
US4440763A (en) * 1981-03-18 1984-04-03 Block Drug Company, Inc. Use of 4-aminosalicyclic acid as an anti-inflammatory agent
US4751221A (en) * 1985-10-18 1988-06-14 Sloan-Kettering Institute For Cancer Research 2-fluoro-arabinofuranosyl purine nucleosides
US5013727A (en) * 1980-03-20 1991-05-07 Farmaceutisk Laboratorium Ferring A/S Pharmaceutical composition and method for the treatment of colitis ulcerosa and Crohn's disease by oral administration
US5245080A (en) * 1989-02-20 1993-09-14 Jouveinal Sa (+)-1-[(3,4,5-trimethoxy)-benzyloxymethyl]-1-phenyl-N,N-dimethyl-N-propylamine, process for preparing it and its therapeutical use
US5541170A (en) * 1981-07-31 1996-07-30 Tillotts Pharma Ag Orally administrable pharmaceutical compositions
US5811547A (en) * 1992-10-14 1998-09-22 Nippon Shinyaju Co., Ltd. Method for inducing crystalline state transition in medicinal substance
US6197341B1 (en) * 1997-03-14 2001-03-06 Stefan Friess Formulations of balsalazide and its derivatives
US6353024B1 (en) * 1999-12-23 2002-03-05 Warner-Lambert Company Method for preventing and treating arthritis, osteo-traumatic pain, and neuralgias with trimebutine
US6458776B1 (en) * 2000-08-29 2002-10-01 Nobex Corporation 5-ASA derivatives having anti-inflammatory and antibiotic activity and methods of treating diseases therewith
US20030119903A1 (en) * 1999-12-23 2003-06-26 Agnes Grouhel Use of trimebutine for treating pain
US6602915B2 (en) * 2000-07-27 2003-08-05 Rutgers, The State University Of New Jersey Therapeutic azo-compounds for drug delivery
US20030166630A1 (en) * 2000-05-05 2003-09-04 Serge Auvin Amino acid derivatives and their use as medicines
US6986901B2 (en) * 2002-07-15 2006-01-17 Warner-Lambert Company Llc Gastrointestinal compositions
US7041704B2 (en) * 2003-01-30 2006-05-09 Dynogen Pharmaceuticals, Inc. Methods of treating gastrointestinal tract disorders using sodium channel modulators

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997009329A1 (en) 1995-09-08 1997-03-13 J. Uriach & Cia. S.A. Azo derivatives of 5-aminosalicylic acid for treatment of inflammatory bowel disease
AU7246398A (en) * 1997-04-30 1998-11-24 Northwestern University Inhibition of nitric oxide synthase by amino acids and dipeptides
NZ537395A (en) * 2002-07-10 2006-11-30 Warner Lambert Co Combination of an allosteric inhibitor or matrix metalloproteinase-13 with a selective inhibitor of cyclooxygenase-2 that is not celecoxib or valdecoxib
EP1886681A3 (en) 2004-10-07 2008-11-19 Sulfidris S.r.l. 5-(p-hydroxyphenyl)-3H-1,2-dithiol-3-thione valproate ester

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4301163A (en) * 1979-07-11 1981-11-17 Societe Industrielle De Produits De Synthese Amino-ether oxides and use thereof in therapy
US4301163B1 (en) * 1979-07-11 1990-01-23 Synthese Soc Ind Prod
US5013727A (en) * 1980-03-20 1991-05-07 Farmaceutisk Laboratorium Ferring A/S Pharmaceutical composition and method for the treatment of colitis ulcerosa and Crohn's disease by oral administration
US4412992A (en) * 1980-07-21 1983-11-01 Biorex Laboratories Limited 2-Hydroxy-5-phenylazobenzoic acid derivatives and method of treating ulcerative colitis therewith
US4440763A (en) * 1981-03-18 1984-04-03 Block Drug Company, Inc. Use of 4-aminosalicyclic acid as an anti-inflammatory agent
US5541170A (en) * 1981-07-31 1996-07-30 Tillotts Pharma Ag Orally administrable pharmaceutical compositions
US4751221A (en) * 1985-10-18 1988-06-14 Sloan-Kettering Institute For Cancer Research 2-fluoro-arabinofuranosyl purine nucleosides
US5245080A (en) * 1989-02-20 1993-09-14 Jouveinal Sa (+)-1-[(3,4,5-trimethoxy)-benzyloxymethyl]-1-phenyl-N,N-dimethyl-N-propylamine, process for preparing it and its therapeutical use
US5811547A (en) * 1992-10-14 1998-09-22 Nippon Shinyaju Co., Ltd. Method for inducing crystalline state transition in medicinal substance
US6197341B1 (en) * 1997-03-14 2001-03-06 Stefan Friess Formulations of balsalazide and its derivatives
US6353024B1 (en) * 1999-12-23 2002-03-05 Warner-Lambert Company Method for preventing and treating arthritis, osteo-traumatic pain, and neuralgias with trimebutine
US20020165276A1 (en) * 1999-12-23 2002-11-07 Gilles Brunelle Method for preventing and treating inflammatory somatic pain and chronic pain
US20030119903A1 (en) * 1999-12-23 2003-06-26 Agnes Grouhel Use of trimebutine for treating pain
US20030166630A1 (en) * 2000-05-05 2003-09-04 Serge Auvin Amino acid derivatives and their use as medicines
US6602915B2 (en) * 2000-07-27 2003-08-05 Rutgers, The State University Of New Jersey Therapeutic azo-compounds for drug delivery
US6458776B1 (en) * 2000-08-29 2002-10-01 Nobex Corporation 5-ASA derivatives having anti-inflammatory and antibiotic activity and methods of treating diseases therewith
US6986901B2 (en) * 2002-07-15 2006-01-17 Warner-Lambert Company Llc Gastrointestinal compositions
US7041704B2 (en) * 2003-01-30 2006-05-09 Dynogen Pharmaceuticals, Inc. Methods of treating gastrointestinal tract disorders using sodium channel modulators

Also Published As

Publication number Publication date
EP2024323B1 (en) 2013-03-27
EP2024323A4 (en) 2010-07-28
RU2008152751A (en) 2010-07-20
JP2009539777A (en) 2009-11-19
EP2024323A1 (en) 2009-02-18
KR20090021346A (en) 2009-03-03
CA2653869A1 (en) 2007-12-13
JP5421098B2 (en) 2014-02-19
WO2007140611A1 (en) 2007-12-13
US7666907B2 (en) 2010-02-23
BRPI0712417A2 (en) 2012-09-04
AU2007257174B2 (en) 2012-07-12
AU2007257174A1 (en) 2007-12-13
CN101489986A (en) 2009-07-22
US20070275905A1 (en) 2007-11-29

Similar Documents

Publication Publication Date Title
US7666907B2 (en) Salts of trimebutine and N-desmethyl trimebutine
JP5221341B2 (en) 4- or 5-aminosalicylic acid derivatives
EP2041108B1 (en) 4-hydroxythiobenzamide derivatives of drugs
US20060270635A1 (en) Derivatives of 4- or 5-aminosalicylic acid
IE49701B1 (en) Amides of acyl-carnitines,process for preparing same and pharmaceutical compositions containing such amides
US8114858B2 (en) Derivatives of 4- or 5-aminosalicylic acid
AU2014276346A1 (en) Compositions and methods for the treatment of diabetes and pre-diabetes
US9642915B2 (en) Compositions and methods for the treatment of neuromuscular disorders and neurodegenerative diseases
WO2014174425A2 (en) Compositions and methods for the treatment of orthostasis and neurological diseases
US9108942B1 (en) Compositions and methods for the treatment of moderate to severe pain
EP3481383B1 (en) Compositions and methods for the treatment of irritable bowel syndrome
PT1043332E (en) Dipeptides comprising cyclopentan-beta-amino acids
US20110237658A1 (en) Compound and method for the treatment of pain
WO2014068461A2 (en) Compositions and methods for the treatment of acute inflammation
NO170413B (en) ANALOGY PROCEDURE FOR THE PREPARATION OF NEW, THERAPEUTIC-ACTIVE CYSTEIN DERIVATIVES
WO2014147531A2 (en) Compositions and methods for the treatment of cancer
WO2015022613A1 (en) Compositions and methods for the treatment of diabetes and pre-diabetes
WO2014191885A2 (en) Compositions and methods for the treatment of neuromuscular disorders
WO2014091467A2 (en) Compositions and methods for the treatment of autoimmune diseases and inflammation
US20150119424A1 (en) Compositions and methods for the treatment of epilepsy and neurological diseases
WO2015001499A2 (en) Compositions and methods for the treatment of irritable bowel syndrome and the associated abdominal cramping
WO2015028928A2 (en) Compositions and methods for the treatment of inflammation and arthritis
WO2013168007A1 (en) Compositions and methods for the treatment of cardiovascular and neurological diseases

Legal Events

Date Code Title Description
AS Assignment

Owner name: ANTIBE THERAPEUTICS INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WALLACE, JOHN L.;CIRINO, GIUSEPPE;SANTAGADA, VINCENZO;AND OTHERS;SIGNING DATES FROM 20070611 TO 20070612;REEL/FRAME:021907/0463

AS Assignment

Owner name: GICARE PHARMA INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ANTIBE HOLDINGS INC.;REEL/FRAME:026765/0789

Effective date: 20110719

Owner name: ANTIBE HOLDINGS INC., CANADA

Free format text: CHANGE OF NAME;ASSIGNOR:ANTIBE THERAPEUTICS INC.;REEL/FRAME:026766/0253

Effective date: 20091211

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION