WO2012114342A1 - Flumazenil complexes, compositions comprising same and uses thereof - Google Patents

Flumazenil complexes, compositions comprising same and uses thereof Download PDF

Info

Publication number
WO2012114342A1
WO2012114342A1 PCT/IL2012/050060 IL2012050060W WO2012114342A1 WO 2012114342 A1 WO2012114342 A1 WO 2012114342A1 IL 2012050060 W IL2012050060 W IL 2012050060W WO 2012114342 A1 WO2012114342 A1 WO 2012114342A1
Authority
WO
WIPO (PCT)
Prior art keywords
flumazenil
sodium
pharmaceutical composition
acid
agent
Prior art date
Application number
PCT/IL2012/050060
Other languages
French (fr)
Inventor
Shimon Amselem
Original Assignee
Coeruleus Ltd.
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 Coeruleus Ltd. filed Critical Coeruleus Ltd.
Priority to CA2827336A priority Critical patent/CA2827336C/en
Priority to RU2013139247/04A priority patent/RU2013139247A/en
Priority to CN201280019999.3A priority patent/CN103502224A/en
Priority to ES12749603T priority patent/ES2702848T3/en
Priority to EP12749603.2A priority patent/EP2678320B1/en
Priority to AU2012221704A priority patent/AU2012221704B2/en
Priority to JP2013554977A priority patent/JP2014511382A/en
Publication of WO2012114342A1 publication Critical patent/WO2012114342A1/en
Priority to IL227742A priority patent/IL227742A/en
Priority to US13/974,760 priority patent/US9585893B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • A61K31/55171,4-Benzodiazepines, e.g. diazepam or clozapine condensed with five-membered rings having nitrogen as a ring hetero atom, e.g. imidazobenzodiazepines, triazolam
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/455Nicotinic acids, e.g. niacin; Derivatives thereof, e.g. esters, amides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/40Cyclodextrins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/0056Mouth soluble or dispersible forms; Suckable, eatable, chewable coherent forms; Forms rapidly disintegrating in the mouth; Lozenges; Lollipops; Bite capsules; Baked products; Baits or other oral forms for animals
    • 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
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/26Androgens

Definitions

  • the present invention provides soluble complexes of flumazenil, methods for the preparation thereof, pharmaceutical compositions comprising same and use of said compositions for alleviating or counteracting the various types of hypersomnia, drowsiness, residual effects associated with the administration of sleep/hypnotic drugs, sedation, alcohol intoxication and hepatic encephalopathy.
  • Flumazenil (chemically named ethyl 8-fluoro- 5,6-dihydro- 5-methyl- 6-oxo- 4H- imidazo [1,5-a] [1,4] benzodiazepine- 3-carboxylate), also known as, Ro 15-1788, AnexateTM, LanexatTM, MaziconTM and RomaziconTM, was initially disclosed in US 4,316,839. Flumazenil is an imidazobenzodiazepine having high affinity for the GAB A A /benzodiazepine-receptor complex, the specific binding site of benzodiazepines.
  • flumazenil is a competitive inhibitor of benzodiazepines, it is used for reversing benzodiazepine-induced sedation and anesthesia following therapeutic or diagnostic procedures (e.g. WO 2009/114740). Flumazenil is also known to reverse the effect of non- benzodiazepine drugs, such as the imidazopyridine hypnotic Zolpidem (e.g. Patat et al, Clin Pharmacol Then, 1994, 56(4):430-6). Flumazenil in combination with hydroxyzine and gabapentin are used for treating stimulant addiction (methamphetamine) (Bond AT, 1998, CNS Drugs, 9(1): 41-57).
  • Flumazenil is also effective in treating hepatic encephalopathy (Als-Nielsen B et al. Cochrane Database of Systematic Reviews 2004, Issue 2. Art. No. : CD002798. DOI: 10.1002/14651858. CD002798.pub2). The aforementioned therapeutic effects were achieved by intravenous administration of flumazenil in a liquid formulation.
  • a liquid formulation of flumazenil (Romazicon®) is currently approved for reversing the sedative effects of benzodiazepines. Due to the limited solubility of flumazenil, the Romazicon® liquid formulation contains only 0.01% flumazenil. Poor water solubility frequently correlates with low drug absorption and bioavailability, and limits the amount of drug that can be administered in a pharmaceutical composition.
  • hydrotopes such as nicotinamide
  • insoluble compounds in order to improve their solubility
  • the present invention is directed to pharmaceutically acceptable complexes of flumazenil, comprising flumazenil and a complexation agent.
  • the flumazenil complexes of the invention are highly soluble while maintaining the therapeutic activity known for flumazenil, namely, ameliorating or inhibiting any type of hypersomnia and drowsiness, including endogenous hypersomnia associated with GABA hyper-activation or exogenic hypersomnia e,g, residual hypersomnia associated with the administration of sleep/hypnotic drugs and ameliorating or inhibiting alcohol intoxication, or treating hepatic encephalopathy or any other disease or disorder associated with GABA related sedation or hypnotics.
  • the flumazenil complexes of the invention offer a pronounced advantage over flumazenil compositions currently known in the art. Moreover, the flumazenil complexes of invention are advantageous for the preparation of formulations suitable for self-administration, such as, sublingual administration, thereby avoiding the involvement of professional care takers and the need of admitting to a health center.
  • the present invention is based in part on the unexpected discovery that flumazenil complexes are more soluble than flumazenil alone. Accordingly, these compounds are suitable for the preparation of highly concentrated pharmaceutical compositions of flumazenil. This enables administration of higher doses of flumazenil (per volume) than currently available.
  • the concentration of flumazenil in the form of the flumazenil complexes of the invention is within the range of about 0.4 to 2%. This concentration is much higher than the concentration of flumazenil in commercially available formulations. In fact, the concentration of flumazenil in the pharmaceutical compositions of the invention is higher by more than two orders of magnitude from the concentration of flumazenil in the commercially available formulations.
  • Highly concentrated flumazenil compositions are particularly advantageous in view of the maximal recommended daily dose approved for flumazenil solution (for example, Romazicon®), which is 3 mg.
  • flumazenil solution for example, Romazicon®
  • concentration of flumazenil in the commercial solution is only 0.01%
  • a dose of 3 mg requires administration of a large volume of the flumazenil solution, as high as 30 ml.
  • the present invention overcomes this deficiency by providing concentrated formulations that can be administered in lower volumes thus improving patient compliance.
  • Administering highly concentrated formulations of flumazenil also enables applying more routes of delivery, specifically, sublingual, submucosal, intramuscular, transdermal or any parentral route of administration.
  • Sublingual administration of the formulations of the invention is of special advantage. Apart from being easy to administer, the sublingual route bypasses the first liver metabolism, absorbance of flumazenil is immediate, thereby fast response to therapy can be achieved.
  • the formulations of the invention are stable and no precipitation occurs during storage.
  • the stability of the formulations of the invention renders them suitable for long periods of storage.
  • the present invention provides a complex of ethyl 8-fluoro- 5,6-dihydro- 5-methyl- 6-oxo- 4H- imidazo [1,5-a] [1,4] benzodiazepine- 3-carboxylate (flumazenil) or a salt thereof and a complexation agent or derivatives thereof, with the proviso that the complexation agent is other than cyclodextrin or meglumine.
  • the present invention provides a complex of ethyl 8- fluoro- 5,6-dihydro- 5-methyl- 6-oxo- 4H- imidazo [1,5-a] [1,4] benzodiazepine- 3- carboxylate (flumazenil) or a salt thereof and an aromatic stacking complexation agent.
  • the stacking complexation agent is selected from the group consisting of: benzoic acid, benzoate, hydroxybenzoic acid, hydroxybenzoate, salicylic acid, salicylate, nicotinamide, nicotinic acid, nicotinate, gentisic acid, gentisate, ethanolamide, toluic acid (ortho, meta or para), toluate (ortho, meta or para), aminobenzoic acid, aminobenzoate, anthranilic acid, anthranilate, butylmonoglycolsulfate and resorcinol.
  • the stacking complexation agent is selected from the group consisting of: benzoic acid, sodium benzoate, potassium benzoate, hydroxybenzoic acid, sodium hydroxybenzoate, potassium hydroxybenzoate, salicylic acid, sodium salicylate, potassium salicylate, nicotinamide, nicotinic acid, sodium nicotinate, potassium nicotinate, gentisic acid, sodium gentisate, potassium gentisate, ethanolamide, sodium toluate, toluic acid, potassium toluate, aminobenzoic acid, sodium aminobenzoate, potassium aminobenzoate, anthranilic acid, sodium anthranilate, potassium anthranilate, sodium butylmonoglycolsulfate, potassium butylmonoglycolsulfate and resorcinol.
  • the stacking complexation agent is selected from the group consisting of: sodium benzoate, sodium hydroxybenzoate, sodium salicylate, nicotinamide, sodium nicotinate, sodium gentisate, gentisic acid ethanolamide, sodium toluate, sodium aminobenzoates, sodium anthranilate, sodium butylmonoglycolsulfate and resorcinol.
  • the complexation agent of the flumazenil complex of the current invention may be any complexation agent known in the art other than cyclodextrin.
  • the present invention contemplates compositions comprising the flumazenil complexes of the invention, and cyclodextrin as an additional excipient.
  • the complexation agent is nicotinamide or a derivative thereof.
  • the nicotinamide derivative is selected from the group consisting of: 2-amino-nicotinamide derivatives, 5 -phenyl-nicotinamide derivative and 6-substituted nicotinamide derivative.
  • the complexation agent: flumazenil ratio is in the range of 1 : 1 to 2: 1.
  • the present invention provides a flumazenil nicotinamide stacking complex.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the flumazenil complex and a pharmaceutical acceptable carrier.
  • the pharmaceutical composition is in a form selected from the group consisting of: a pill, tablet, lozenge, coated tablet, granule, capsule, hard or soft gelatin capsule, aqueous solution, alcoholic solution, oily solution, syrup, emulsion suspension pastille suppository, solution for injection or infusion, ointment, tincture, cream, lotion, powder, spray, transdermal therapeutic systems, nasal spray, buccal spray, aerosol mixture, microcapsule, implant, rod and plaster.
  • a pill tablet, lozenge, coated tablet, granule, capsule, hard or soft gelatin capsule, aqueous solution, alcoholic solution, oily solution, syrup, emulsion suspension pastille suppository, solution for injection or infusion, ointment, tincture, cream, lotion, powder, spray, transdermal therapeutic systems, nasal spray, buccal spray, aerosol mixture, microcapsule, implant, rod and plaster.
  • the pharmaceutical composition is in a form selected from the group consisting of: immediate release, delayed release, pulsatile release, continuous release and repetitive release.
  • immediate release delayed release
  • pulsatile release continuous release
  • repetitive release continuous release
  • the composition is in a solid dosage form. According to yet another embodiment, the composition is suitable for oral administration.
  • flumazenil concentration in the pharmaceutical composition is within the range of about 0.4 to 2 % w/w. According to yet another embodiment, the concentration of flumazenil is within the range of about 0.5 to 1.8 % w/w. According to certain embodiments, the concentration of flumazenil is within the range of about 0.6 to 1.5 % w/w.
  • the pharmaceutical composition comprises flumazenil complex as an active ingredient, and a solubilizing agent selected from an alcohol, a glycol and a combination thereof.
  • a solubilizing agent selected from an alcohol, a glycol and a combination thereof.
  • the solubilizing agent comprises a combination of an alcohol and a glycol wherein the alcohol: glycol ratio is at least 1.5: 1. According to yet another embodiment, the alcohol: glycol ratio is in the range of 1.5: 1 to 5: 1.
  • the solubilizing agent comprises a combination of an alcohol and a glycol wherein the concentration of the solubilizing agent is at least 40%. According to yet another embodiment, the solubilizing agent comprises a combination of an alcohol and a glycol wherein the concentration of the solubilizing agent is in the range of 40% to 60%). According to yet another embodiment, the solubilizing agent comprises ethanol and propylene glycol.
  • the pharmaceutical composition further comprises a buffering agent.
  • the pharmaceutical composition further comprises at least one agent selected from the group consisting of: a penetration enhancer, a surfactant and cyclodextrin, as an inclusion complexing agent.
  • a penetration enhancer e.g., a surfactant, cyclodextrin, as an inclusion complexing agent.
  • the cyclodextrin is hydroxypropyl ⁇ - cyclodextrin (HPCD).
  • HPCD hydroxypropyl ⁇ - cyclodextrin
  • the cyclodextrin is preferably formulated in a buffer having a pH from about 3 to about 6.
  • the cyclodextrin e.g., HPCD
  • the citric acid buffer having a pH of about 4.
  • the preservative is selected from the group consisting of benzyl alcohol, propylparaben, methylparaben and combinations thereof.
  • the preservative is benzyl alcohol.
  • the preservative is a combination of propylparaben and methylparaben.
  • the penetration enhancer is menthol.
  • the buffering agent is selected from the group consisting of: citric buffer, sodium chloride and combination thereof.
  • citric buffer sodium chloride and combination thereof.
  • the surfactant is a cationic surfactant. According to yet another embodiment, the surfactant is benzalkonium chloride.
  • the present invention provides a method for treating a disease or disorder associated with binding of an agent to the GABA receptor, comprising administering to a patient in need thereof a pharmaceutical composition comprising as an active ingredient a flumazenil complex, and at least one pharmaceutical acceptable carrier.
  • treating a disease or disorder associated with binding of an agent to the GABA receptor comprises reversing the effect caused by the binding of the agent to the GABA receptor.
  • the agent is a benzodiazepine a non-benzodiazepine the binding of which to the GABA receptor causes sedation.
  • the present invention provides a method for treating a disease or disorder comprising administering to a patient in need thereof a pharmaceutical composition comprising as an active ingredient a flumazenil complex, and at least one pharmaceutical acceptable carrier, wherein the disease or disorder is selected from the group consisting of: excessive sleepiness, alcohol intoxication and hepatic encephalopathy.
  • the present invention provides a method for treating excessive sleepiness comprising administering to a patient in need thereof a pharmaceutical composition comprising as an active ingredient a flumazenil complex, and at least one pharmaceutical acceptable carrier.
  • the excessive sleepiness is selected from the group consisting of: excessive sleepiness associated with hypersomnia, excessive sleepiness associated with drowsiness, symptoms associated with overdose of a sleep drug, excessive sleepiness associated with sedation and anesthesia induced by a sleep drug, excessive sleepiness associated with sleep drug addiction, excessive sleepiness associated with stimulant addiction, Alzheimer's disease, anxiety, schizophrenia, specifically, schizophrenia associated with excessive sleepiness, the rebound effect of a sleep drug, balance impairment induced by a sleep drug or any combination thereof.
  • the excessive sleepiness is caused by alcohol intoxication.
  • the stimulant is methamphetamine.
  • treating excessive sleepiness associated with stimulant addiction further comprises administering the flumazenil complex, or the pharmaceutical composition comprising same, in combination with hydroxyzine and gabapentin.
  • the sleep-drug is selected from the group consisting of a benzodiazepine, a benzodiazepine modulator, a benzodiazepine analog, a non- benzodiazepine, a 5-HT2A receptor antagonist, a melatonin receptor agonist, an orexin receptor antagonist, a selective serotonin reuptake inhibitor (SSRI), an antihistamine and an herbal product.
  • a benzodiazepine a benzodiazepine modulator, a benzodiazepine analog, a non- benzodiazepine, a 5-HT2A receptor antagonist, a melatonin receptor agonist, an orexin receptor antagonist, a selective serotonin reuptake inhibitor (SSRI), an antihistamine and an herbal product.
  • SSRI selective serotonin reuptake inhibitor
  • the sleep drug is a sedative/hypnotic drug.
  • the sleep drug is a benzodiazepine sedative/hypnotic drug.
  • the benzodiazepine sleep drug is selected from the group consisting of alprazolam, bromazepam, clonazepam, clotiazepam, cloxazolam, diazepam, estazolam, etizolam, fludiazepam, flunitrazepam, flurazepam, halazepam, haloxazolam, lorazepam, medazepam, midazolam, nimetazepam, nitrazepam, olanzapine, oxazepam, quazepam, temazepam and triazolam.
  • treating excessive sleepiness comprises reversing benzodiazepine-induced sedation and anesthesia following therapeutic or diagnostic procedures.
  • the sleep-drug is a non-benzodiazepine sedative/hypnotic drug.
  • the non-benzodiazepine sleep drug is selected from the group consisting of adipiplon (NG-2-73), agomelatine, almoxerant (ACT-078573), brotizolam, diphenhydramine, divaplon, doxepin, eplivanserin (SR 46349), doxylamine succinate, eszopiclone, indiplon, ocinaplon, pagoclone, pazinaclone, pruvanserin (EMD 281014), suproclone, suriclone, L-tryptophan, 5-hydroxy-L-tryptophan, melatonin, melatonin receptor agonists, such as VEC-162 and PD-6735, muramyl dipeptide, ramelteon, sleep-promoting substance, uridine, volinanserin
  • the present invention provides a method for treating alcohol intoxication comprising administering to a patient in need thereof a pharmaceutical composition comprising the flumazenil complex of the invention, and at least one pharmaceutical acceptable carrier.
  • the excessive sleepiness caused by alcohol intoxication is treated by any one or more of reversing the effects of alcohol intoxication, reducing the effects of alcohol intoxication, alleviating the effects of alcohol intoxication and/or improving performance after alcohol consumption.
  • the present invention provides a method for treating hepatic encephalopathy, comprising administering to a subject in need thereof the flumazenil complex of the invention or a pharmaceutical composition comprising same.
  • the route of administration is selected from the group consisting of: oral, buccal, sublingual, transdermal, transmucosal, intranasal, intravenous ( .v.), intraperitoneal (i.p.), intramuscular (i.m.), subcutaneous (s.c.) or intrathecal (i. t.).
  • oral, buccal, sublingual, transdermal, transmucosal, intranasal intravenous ( .v.), intraperitoneal (i.p.), intramuscular (i.m.), subcutaneous (s.c.) or intrathecal (i. t.).
  • the methods of the invention further comprise co-administering, concomitantly or in sequence, a pharmaceutical composition comprising a flumazenil complex together with a wakefulness promoting agent.
  • the wakefulness promoting agent is selected from the group consisting of modafinil, armodafinil, adrafinil, methylphenidate, nefazodone, sodium oxybate, phentermine, pemoline, adrenaline, methylxantines, theobromine, caffeine and a combination thereof.
  • the present invention provides a flumazenil complex for the treatment of excessive sleepiness or hepatic encephalopathy
  • the flumazenil complex comprises flumazenil and a complexation agent, with the proviso that the complexation agent is other than cyclodextrin or meglumine.
  • the present invention provides a flumazenil complex for the treatment of excessive sleepiness or hepatic encephalopathy
  • the flumazenil complex comprises flumazenil and an aromatic complexation agent, such as, nicotinamide derivatives.
  • the present invention provides a pharmaceutical composition comprising a flumazenil complex as the active ingredient, for the treatment of excessive sleepiness or hepatic encephalopathy.
  • Figure 1 is a schematic presentation of a flumazenil hydrochloric acid salt.
  • Figure 2 exhibits a chromatogram of 0.6 mg/ml nicotinamide solution in diluents.
  • Figure 3 shows a chromatogram of 40 ⁇ g/ml flumazenil solution in diluents.
  • Figure 4 shows a chromatogram of a flumazenil nicotinamide complex in diluents.
  • Figure 5 presents the body weight distribution of the rats (lF-treatment and 2F- placebo) in the study of reversal diazepam-induced sedative-hypnotic effects.
  • Figure 6 exhibits the sleeping time of the rats (lF-treatment and 2F-placebo) following treatment.
  • the present invention is directed to pharmaceutically acceptable flumazenil salts comprising flumazenil and a counter-ion.
  • the present invention provides pharmaceutically acceptable flumazenil complexes comprising flumazenil with a stacking complexation agent, such as an aromatic complexation agent, wherein the complexation agent is other than meglumine and cyclodextrin.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • examples of pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid salts of basic residues such as amines and the like.
  • the pharmaceutically acceptable salts include conventional non-toxic salts of the parent compound formed, for example, from inorganic or organic acids, such as mineral acids (e.g., HC1) and nicotinic acids.
  • non-toxic salts include those derived from inorganic acids, such as, hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids, such as, acetic, propionic, succinic, glycolic, stearic, lactic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
  • inorganic acids such as, hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like
  • organic acids such as, acetic, propionic, succinic, glycolic, stearic, lactic
  • the salts of the present invention are acid addition salts of flumazenil. According to some embodiments, the salts of the present invention are acid addition salts of flumazenil other than the acid addition salt flumazenil HC1.
  • Acid addition salts may, for example, be formed by mixing a solution of the compound of the present invention with a solution of a pharmaceutically acceptable acid, such as, hydrochloric acid, sulfuric acid, acetic acid, trifluoroacetic acid and benzoic acid.
  • a pharmaceutically acceptable acid such as, hydrochloric acid, sulfuric acid, acetic acid, trifluoroacetic acid and benzoic acid.
  • acid salt and “acid addition salts” are synonymous and refer to a class of salts formed by the partial neutralization of protic, diprotic or polyprotic acids. In diprotic or polyprotic acids, because the parent acid is only partially neutralized, one or more replaceable protons remain. Acid salt compounds can act either as an acid or a base: addition of a suitably strong acid will restore protons, and addition of a suitably strong base will remove protons. The pH of a solution of an acid salt will depend on the relevant equilibrium constants and the amounts of any additional base or acid.
  • the flumazenil complex of the invention comprises flumazenil and a complexation agent, wherein the complexation agent is preferably a hydrotope, other than cyclodextrins or meglumine, preferably, an aromatic stacking complexation agent.
  • hydrotopes refers to compounds which are capable of opening up the water structure associated with hydrophobic (lipophilic) and other molecules. These compounds may be used to enhance the aqueous solubility of poorly water-soluble substances.
  • hydrotopes includes sodium benzoate, sodium hydroxybenzoate, sodium salicylate, nicotinamide, sodium nicotinate, urea, sodium gentisate, gentisic acid ethanolamide, sodium toluate, sodium aminobenzoates, sodium anthranilate, sodium butylmonoglycolsulfate, resorcinol and the like. Each possibility represents a separate embodiment of the present invention.
  • stacking complexation agent refers to complexation agent that forms a layered structure consisting of planar layers of the complexation agent stacked above and below the compounds with which the complex is formed. This mechanism is particularly suitable for complexing insoluble molecules having a relatively planar configuration, such as flumazenil. Preferably, the stacking complexation agent also has a relatively planar configuration.
  • Preferred stacking complexation agents include nicotinamide and nicotinamide derivatives.
  • nicotinamide derivatives as used herein is meant to include any derivative of nicotinamide capable of forming a complex with flumazenil with improved solubility compared to the solubility of uncomplexed flumazenil.
  • Acceptable nicotinamide derivatives include, but are not limited to, the nicotinamide derivatives disclosed in any one or more of: US Patent Nos. 7,056,934 and 7, 153,870; WO 2008/071665; Kuramochia et al, Bioorganic & Medicinal Chemistry, Vol. 13(12), 2005, 4022-4036; and Fonti et al, Nucl Med Biol, 1999, 26(6): 681-6, among others.
  • nicotinamide derivatives include 2-amino-nicotinamide derivatives, 5 -phenyl-nicotinamide derivatives, and 6- substituted nicotinamide derivatives.
  • Complex formation which is non-covalent in nature, may be achieved by mixing appropriate ratios of the solvent-derived composition and the hydrotope or mixtures thereof in a suitable liquid vehicle, which may be aqueous, organic or a combination of both. Additional excipients, such as, surfactants, polyols, disaccharides may be added to facilitate complexation or for assisting in dispersability.
  • the resultant complex may be isolated as a dry powder by any process known in the art (co-precipitation and drying, evaporation of the liquid vehicle, spray drying, lyophilization etc.). Particle size may be reduced by any standard technique known in the art, if desired.
  • the resultant hydrotope complex may be used without further modification or may be compounded into a variety of other formulations or vehicles as required.
  • the flumazenil stacking complex of the invention is a flumazenil nicotinamide complex.
  • Nicotinamide also known as niacinamide and nicotinic acid amide, is the amide of nicotinic acid (vitamin B3/niacin) and is also known as 3- pyridinecarboxamide, nicotinic acid amide and Vitamin PP.
  • Nicotinamide has over the past years been given at high doses for a variety of therapeutic applications.
  • nicotinamide is classed as a food additive rather than a drug, a category that does require the formal safety evaluation normally expected of new drugs and therapies.
  • the therapeutic index of nicotinamide is wide but at very high doses (i.e. in megadoses) reversible hepatotoxicity has been reported in animals and humans. Minor abnormalities of liver enzymes can infrequently occur at the doses used for diabetes prevention. There is no evidence of teratogenicity from animal studies and there is no evidence of oncogenicity in human. Growth inhibition was shown in rats but growth in children is unaffected.
  • nicotinamide In addition, minor degrees of insulin resistance attributed to nicotinamide have been reported. High-dose nicotinamide, i.e. in excess of 3 gm/day, should be considered as a drug with toxic potential at adult doses and unsupervised use should be discouraged.
  • the ratio of flumazenil: nicotinamide in the stacking complex of the invention is within the range of 1 : 1 to 1 :2. In certain embodiments the flumazenil: nicotinamide ratio in the complex of the invention is about 1 : 1.5. This ratio increases the solubility of flumazenil such that formulations comprising about 1% flumazenil with excellent solubility are obtained. The amount of nicotinamide in such formulations is about 1.5%. Accordingly, for example, a pharmaceutical composition comprising 3 mg of flumazenil, which is about the maximal daily dose allowed for flumazenil, corresponds to 4.5 mg of nicotinamide. An amount of 4.5 mg nicotinamide is lower by a factor of about 670 from 3 gr. (where 3 gr. is considered high dose and potentially toxic, for nicotinamide).
  • cyclodextrins another family of complexation agents, which increase the solubility of poorly soluble compounds by means of inclusion, are found toxic at relatively low concentrations, although there are significant differences between the cytotoxicity of the various cyclodextrin derivatives.
  • solubilized paclitaxel (as Taxol®), a poorly soluble drug, increased with cyclodextrin concentration.
  • some of the cyclodextrins tested in this study were found toxic in mice with a maximum tolerated dose of 2 g/kg body weight, which is the quantity of cyclodextrin required to administer paclitaxel at 10 mg/kg.
  • Other cyclodextrins allowed paclitaxel administration at higher doses yet had a maximum tolerated dose of 25 mg drug/kg.
  • flumazenil-nicotinamide complex of the invention Another advantage attributed to the flumazenil-nicotinamide complex of the invention is long shelf life. As exemplified hereinbelow, the flumazenil complex of the invention has an exceptional stability, during days, weeks and month of storage at room temperature and even at higher temperatures.
  • cyclodextrins and nicotinamide increase solubility by different complexation mechanisms. It has been suggested that nicotinamide complex with hydrophobic ('guest') species is made out of a stack of nicotinamide and the guest species, whereby one or more 'sheets' of nicotinamide are stacked above and below a layer of the hydrophobic species. Clearly, stack formation depends on the 3D configuration of the members of the complex. While nicotinamide is substantially planar, for the formation of a stacked complex, it is generally required that the hydrophobic guest species would also be planar. Complex formation by cyclodextrins is exerted by way of inclusion.
  • Cyclodextrins form a cavity, having an inner hydrophobic core and an outer hydrophilic core, where the cavity traps the hydrophobic species.
  • the resulting complex exhibits modified physical and chemical properties, mostly in terms of water solubility. Cyclodextrins are relatively large molecules (molecular weight ranging from almost 1000 to over 1500), with a hydrated outer surface, and under normal conditions, cyclodextrin molecules will only permeate biological membranes with considerable difficulty.
  • the present invention further provides a pharmaceutical composition comprising the flumazenil salt or complex as the pharmaceutically active ingredient and a pharmaceutical acceptable carrier.
  • the pharmaceutical compositions according to the present invention preferably comprise one or more pharmaceutical acceptable carriers and the active constituent(s).
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • the carriers in the pharmaceutical composition may comprise a binder, such as microcrystalline cellulose, polyvinylpyrrolidone (polyvidone or povidone), gum tragacanth, gelatine, starch, lactose or lactose monohydrate; a disintegrating agent, such as alginic acid, maize starch and the like; a lubricant or surfactant, such as magnesium stearate, or sodium lauryl sulphate; a glidant, such as colloidal silicon dioxide; a sweetening agent, such as sucrose or saccharin; and/or a flavoring agent, such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, polyvinylpyrrolidone (pol
  • the carriers include, but not limited to, physiological saline, polyethylene glycol, ethanol, vegetable oils, isopropyl myristate, etc.
  • the compounds of the present invention can be formulated in the form of ointment or cream. Each possibility represents a separate embodiment of the present invention.
  • the carrier(s) must be “acceptable” in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipients thereof.
  • the pharmaceutical composition comprising the flumazenil salt or complex of the invention as the active ingredient, may further comprise cyclodextrins, at least one solubilizing agent, at least one penetration enhancer, and at least one preservative.
  • the pharmaceutical composition may further comprise additional excipients, such as, flavoring agents, among others.
  • the solubilizing agent is preferably a polar solvent such as mono- or poly-alcohols of linear or branched configuration (e.g., CI to C8 alcohols).
  • a polar solvent such as mono- or poly-alcohols of linear or branched configuration (e.g., CI to C8 alcohols).
  • Non-limiting examples include methanol, ethanol, propanol, iso-propanol, n-butanol, sec-butanol, isobutanol, t-butanol, n- pentanol, 2-pentanol, 3-pentanol, neopentanol, 1-hexanol, 2-hexanol, 3-hexanol, 1-heptanol, 2-heptanol, 1-octanol, as well as any geometrical isomers, enantiomers and diastereomers of any of the foregoing.
  • Suitable polar solvents include glycols such as ethylene glycol, propylene glycol and their polymers having a molecular weight between 400 and 1000.
  • the solubilizing agent is a combination of ethanol and propylene glycol.
  • Preferred solubilizing agents include alcohols, glycols and combinations thereof. Each possibility represents a separate embodiment of the present invention.
  • the solubilizing agent in the composition of the invention comprises a combination of an alcohol and a glycol wherein the alcohol:glycol ratio is at least 1.5: 1 or in the range of 1.5: 1 to 5: 1.
  • the solubilizing agent preferably in the form of combination of an alcohol and a glycol, may be at least 40%, or in the range of 40% to 60%)., of the total weight of the pharmaceutical composition of the invention.
  • cyclodextrin as used herein means ⁇ -, ⁇ - or ⁇ - cyclodextrin or a derivative thereof.
  • Suitable cyclodextrin derivatives for use in the formulations of the present invention include, but are not limited to the cyclodextrin listed above, e.g.
  • hydroxypropyl derivatives of ⁇ -, ⁇ - and ⁇ -cyclodextrin sulfo alky 1 ether cyclodextrins such as sulfobutylether ⁇ -cyclodextrin, alkylated cyclodextrins such as the randomly methylated ⁇ -cyclodextrin, and various branched cyclodextrins such as glucosyl- and maltosyl ⁇ -cyclodextrin.
  • sulfo alky 1 ether cyclodextrins such as sulfobutylether ⁇ -cyclodextrin
  • alkylated cyclodextrins such as the randomly methylated ⁇ -cyclodextrin
  • various branched cyclodextrins such as glucosyl- and maltosyl ⁇ -cyclodextrin.
  • Other cyclodextrins are described in US patent publication US
  • the cyclodextrin is hydroxypropyl ⁇ - cyclodextrin (HPCD).
  • HPCD hydroxypropyl ⁇ - cyclodextrin
  • the cyclodextrin is preferably formulated in a buffer having a pH from about 3 to about 6.
  • the cyclodextrin e.g., HPCD
  • the citric acid buffer having a pH of about 4.
  • the cyclodextrin component of the formulations of the present invention may be present in an amount from about 10%> to about 95% w/w, for example from about 30% to about 80%), from about 30% to about 75%, or about 60% based on the formulations of the invention. It is generally recognized that cyclodextrins act as true carriers by keeping the hydrophobic drug molecules in solution and delivering them to the surface of the biological membrane, e.g. skin, mucosa or the eye cornea, where they partition into the membrane.
  • the relatively lipophilic membrane has low affinity for the hydrophilic cyclodextrin molecules and therefore they remain in the aqueous membrane exterior, e.g. the aqueous vehicle system, salvia or the tear fluid.
  • Conventional penetration enhancers such as alcohols and fatty acids, disrupt the lipid layers of the biological barrier. Cyclodextrins, on the other hand, may act as penetration enhancers by increasing drug availability at the surface of the biological barrier.
  • Therapeutic formulations suitable for oral administration may be obtained by compression or moulding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by mixing the constituent(s), and compressing this mixture in a suitable apparatus into tablets having a suitable size.
  • the phanquinone may be mixed with a binder, a lubricant, an inert diluent and/or a disintegrating agent and the further optionally present constituents may be mixed with a diluent, a lubricant and/or a surfactant.
  • a tablet may be coated or uncoated. An uncoated tablet may be scored.
  • a coated tablet may be coated with sugar, shellac, film or other enteric coating agents.
  • Therapeutic formulations suitable for parenteral administration include sterile solutions or suspensions of the active constituents.
  • An aqueous or oily carrier may be used.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soy bean oil, mineral oil, sesame oil and the like.
  • Formulations for parenteral administration also include a lyophilized powder comprising flumazenil and, optionally, further active constituents, that is to be reconstituted by dissolving in a pharmaceutically acceptable carrier that dissolves the active constituents, e.g. an aqueous solution of carboxymethylcellulose and lauryl sulphate.
  • a pharmaceutically acceptable carrier that dissolves the active constituents
  • the pharmaceutical composition when it is a capsule, it may contain a liquid carrier, such as fatty oil, e.g. cacao butter.
  • a liquid carrier such as fatty oil, e.g. cacao butter.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Each possibility represents a separate embodiment of the present invention.
  • the formulations of the present invention may further comprise penetration enhancers, such as menthol.
  • penetration enhancers that may be used in the formulations of the present invention include, but are not limited to, anionic surfactants (e.g. sodium lauryl sulphate, sodium dodecyl sulphate), cationic surfactants (e.g. palmitoyl DL carnitine chloride, cetylpyridinium chloride), nonionic surfactants (e.g. polysorbate 80, polyoxyethylene 9-lauryl ether, glyceryl monolaurate, polyoxyalkylenes, polyoxyethylene 20 cetyl ether), lipids (e.g. oleic acid), bile salts (e.g.
  • the formulations of the invention optionally further comprise at least one preservative.
  • Any suitable preservative may be present in the formulation in the present invention.
  • the preservative may be any pharmaceutically acceptable preservative, for example methyl 4- hydroxybenzoate (methyl paraben), ethyl 4-hydroxybenzoate (ethyl paraben), propyl 4- hydroxybenzoate (propylparaben), benzyl alcohol, sorbic acid, sodium benzoate, benzoic acid, and any combination thereof.
  • methyl 4- hydroxybenzoate methyl paraben
  • ethyl 4-hydroxybenzoate ethyl paraben
  • propyl 4- hydroxybenzoate propyl 4- hydroxybenzoate
  • benzyl alcohol sorbic acid
  • sodium benzoate benzoic acid
  • benzoic acid and any combination thereof.
  • the formulation of the invention optionally further comprises a flavoring agent in an amount between 0.05 and 10 percent by weight of the total composition.
  • the flavoring agent is present in an amount between 0.1 and 2.5 percent by weight of the total composition.
  • the flavoring agent is preferably selected from the group consisting of synthetic or natural oil of peppermint, oil of spearmint, citrus oil, fruit flavors, sweeteners (sugars, aspartame, saccharin, Estevia, etc.), and mixtures thereof. Menthol can also act as a flavoring agent.
  • the therapeutic compound can be delivered in a controlled release system.
  • controlled release is used herein to refer to a pharmaceutical dosage form in which release of the active ingredient is timed or modified to a rate sufficient to maintain the desired therapeutic level over an extended period of time.
  • the release may be a "sustained release” or a “delayed release” such that release of the active ingredient from the pharmaceutical dosage form is other than promptly after administration of the dosage form, but rather is withheld or delayed following administration.
  • a pump may be used.
  • polymeric materials can be used.
  • a controlled release system can be placed in proximity of the therapeutic target, i.e., the central nervous system, thus requiring only a fraction of the systemic dose.
  • Other controlled release systems are discussed, for example, in U.S. Patent No. 5, 120,548 which is directed a controlled-release drug delivery device comprised of swellable polymers.
  • U.S. Patent No. 5,073,543 also describes controlled-release formulations containing a trophic factor entrapped by a ganglioside-liposome vehicle.
  • 5,639,476 discloses a stable solid controlled-release formulation having a coating derived from an aqueous dispersion of a hydrophobic acrylic polymer.
  • Biodegradable microparticles are also known for use in controlled-release formulations.
  • U.S. Patent No. 5,733,566 describes the use of polymeric microparticles that release antiparasitic compositions.
  • the controlled-release of the active ingredient may be stimulated by various inducers, for example, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • compositions comprising one or more pharmacologically active drugs, and one or more pharmaceutically acceptable excipients, diluents or carriers.
  • compositions, formulations and dosage forms can be designed for administration by all possible administration routes to achieve the desired therapeutic response.
  • the terms used may refer to the physical format of the product which is dispensed and administered to the patient, for example, a capsule or a patch. Alternately or in addition, the terms used may refer to any of: the mode of administration, the mode of delivery or the mode of release of the drug, for example a transdermal delayed release formulation.
  • the compounds of the present invention may be formulated into injections by dissolving, suspending or emulsifying water-soluble solvent such as saline and 5% dextrose, or in water-insoluble solvents such as vegetable oils, synthetic fatty acid glyceride, higher fatty acid esters and propylene glycol.
  • the formulations of the invention may include any of conventional additives such as dissolving agents, isotonic agents, suspending agents, emulsifiers, stabilizers and preservatives.
  • the present invention provides a method for treating a disease or disorder associated with binding of an agent to the GABA receptor, comprising administering to a patient in need thereof a pharmaceutical composition comprising an active ingredient selected from flumazenil salt and flumazenil complex, and at least one pharmaceutical acceptable carrier.
  • a pharmaceutical composition comprising an active ingredient selected from flumazenil salt and flumazenil complex, and at least one pharmaceutical acceptable carrier.
  • the disease or disorder is excessive sleepiness
  • the agent is a benzodiazepine or a non-benzodiazepine, and the effect is sedation.
  • treating encompasses substantially ameliorating, relieving, alleviating and preventing symptoms of a disease, disorder or condition in a subject.
  • administering refers to delivery of a pharmaceutical compound to a subject by any means that does not affect the ability of the compound to perform its intended function.
  • the terms "sleep drug” is used herein in reference to pharmaceutical agents used for inducing and/or maintaining sleep, in particular, prescription sleep drugs that are classified as hypnotics/sedatives.
  • the sleep-drug may be a sedative/hypnotic drug, a benzodiazepine, a benzodiazepine modulator, a benzodiazepine analog, a non-benzodiazepine, a 5-HT2A receptor antagonist, a melatonin receptor agonist, an orexin receptor antagonist, a selective serotonin reuptake inhibitor (SSRI), an antihistamine and an herbal product.
  • SSRI selective serotonin reuptake inhibitor
  • Suitable benzodiazepine sleep drugs include alprazolam, bromazepam, clonazepam, clotiazepam, cloxazolam, diazepam, estazolam, etizolam, fludiazepam, flunitrazepam, flurazepam, halazepam, haloxazolam, lorazepam, medazepam, midazolam, nimetazepam, nitrazepam, olanzapine, oxazepam, quazepam, temazepam and triazolam.
  • Suitable non-benzodiazepine sleep drug include adipiplon, agomelatine, almoxerant, brotizolam, diphenhydramine, divaplon, doxepin, eplivanserin, doxylamine succinate, eszopiclone, indiplon, ocinaplon, pagoclone, pazinaclone, pruvanserin (EMD 281014), suproclone, suriclone, L-tryptophan, 5-hydroxy-L-tryptophan, melatonin, melatonin receptor agonists, muramyl dipeptide, ramelteon, uridine, volinanserin, zaleplon, Zolpidem, imidazopyridine hypnotic Zolpidem, APD125, ACP-103, PD 200-390, HY10275, GW649863 and EVT-201
  • the term "effective amount” refers to an amount of a pharmaceutical compound sufficient to achieve its desired effect.
  • the excessive sleepiness is selected from the group consisting of: excessive sleepiness associated with hypersomnia, excessive sleepiness associated with drowsiness, symptoms associated with overdose of a sleep drug, excessive sleepiness associated with alcohol intoxication, excessive sleepiness associated with sedation and anesthesia induced by a sleep drug, excessive sleepiness associated with sleep drug addiction, excessive sleepiness associated with stimulant addiction, the rebound effect of a sleep drug, balance impairment induced by a sleep drug or any combination thereof.
  • treating excessive sleepiness comprises reversing benzodiazepine-induced sedation and anesthesia following therapeutic or diagnostic procedures.
  • hypothala refers to chronic or recurrent bouts of excessive sleepiness, characterized by one or more of near-daily diurnal sleep episodes, excessive naps, abnormally prolonged sleep intervals, a perception of non-restorative sleep, and difficulty in making the transition from sleep to wakefulness.
  • Hypersomnia may be one or more of: shift work sleep disorder; narcolepsy; obstructive sleep apnea/hypopnea syndrome; REM behavior disorder; frontal nocturnal dystonia; restless legs syndrome; nocturnal movement disorder; Kleine-Levin syndrome; Parkinson's disease; Alzheimer's disease, schizophrenia, anxiety, schizophrenia (in particular, schizophrenia associated with excessive sleepiness), excessive sleepiness; hypersomnia; idiopathic hypersomnia; recurrent hypersomnia; endozepine related recurrent stupor; and amphetamine resistant hypersomnia.
  • the excessive sleepiness is caused by alcohol intoxication.
  • the method of the invention is directed to treating alcohol intoxication.
  • alcohol intoxication means overdose of alcohol (e.g., ethanol) leading to behavioral impairment.
  • alcohol e.g., ethanol
  • a person is said to suffer from alcohol intoxication when the quantity of alcohol the person consumes exceeds the individual's tolerance for alcohol and produces behavioral or physical abnormalities. In other words, the person's mental and physical abilities are impaired.
  • alcohol refers to the generic term for ethanol, which is a particular type of alcohol produced by the fermentation of many foodstuffs— most commonly barley, hops, and grapes.
  • Other types of alcohol commonly available such as methanol (common in glass cleaners), isopropyl alcohol (rubbing alcohol), and ethylene glycol (automobile antifreeze solution) are highly poisonous when swallowed, even in small quantities. Ethanol produces intoxication because of its depressive effects on various areas of the brain causing these impairments in a progressive order as the person gets more and more drunk.
  • Symptoms of alcohol intoxication and/or impaired performance after alcohol consumption include disinhibition of normal social functioning (e.g., excessive talking), loss of memory, confusion, disorientation, uncoordinated movement, progressive lethargy, coma, or ultimately death.
  • treating excessive sleepiness caused by alcohol intoxication is selected from the group consisting of: reversing the effects of alcohol intoxication, reducing the effects of alcohol intoxication, alleviating the effects of alcohol intoxication and improving performance after alcohol consumption.
  • the method of the invention is directed treating hepatic encephalopathy with the flumazenil formulation.
  • the method of the invention provides an improved treatment of hepatic encephalopathy.
  • the pharmaceutical compositions of the present invention are suitable for oral, sublingual or transdermal administration.
  • the pharmaceutical composition of the present invention includes much higher concentrations of flumazenil, of about 0.4 to 2%. This concentration is higher by two orders of magnitude from the concentration of flumazenil in the commercially available flumazenil formulations that are known to date.
  • the pharmaceutical compositions and methods of the present invention provide advantageous patient compliance.
  • the present invention provides attractive routes for the delivery of flumazenil (e.g. sub-mucosaland and transdermal) thereby averting the need for admission to health centers.
  • Hepatic encephalopathy refers to a complex neuropsychiatric syndrome, which may complicate acute or chronic hepatic failure. It is characterized by changes in mental state including a wide range of neuropsychiatric symptoms ranging from minor not readily discernible signs of altered brain function, overt psychiatric and/or neurological symptoms to deep coma.
  • Hepatic encephalopathy occurs in patients with cirrhosis, often associated with spontaneous or iatrogenic portal-systemic shunting. Hepatic encephalopathy is generally considered a reversible metabolic encephalopathy. Traditionally, hepatic encephalopathy has been considered to be secondary to the accumulation of toxic products, which have not been metabolized by the liver.
  • GABA gamma-amino butyric acid
  • liver failure leads to the accumulation of substances that bind to the GABA/benzodiazepine complex resulting in neural inhibition which may progress to coma.
  • a benzodiazepine-receptor antagonist, flumazenil has been assessed in the treatment of hepatic encephalopathy in the hope of reversing neuropsychiatric symptoms related to the accumulation of endogenous benzodiazepine.
  • Als-Nielsen et al. disclose the use of flumazenil, administered via intravenous infusion of 1 mg flumazenil in 20 ml saline solution (0.005%) over 3-5 minutes, for treating hepatic encephalopathy.
  • Als-Nielsen reports that flumazenil had a significant beneficial effect on short-term improvement of hepatic encephalopathy in patients with cirrhosis and a highly favorable prognosis but had no significant effect on recovery or survival.
  • the methods of the invention may be used in conjunction with insomnia treatment modalities, and serve to eliminate or diminish residual soporific effects associated with administration of sleep drugs.
  • the invention is effective for counteracting excessive sleepiness induced by a wide variety of sleep drugs.
  • sleep drugs include benzodiazepine and non-benzodiazepine drugs which are classified as hypnotics/sedatives, as well as other prescription and non-prescription sleep drugs, including those classified as 5-HT 2A receptor antagonists, melatonin receptor agonists, orexin receptor antagonists, selective serotonin reuptake inhibitors (SSRIs), and other sleep inducing agents such as antihistamines, melatonin and certain herbal products. It is to be specifically understood that a particular sleep drug may be classified under more than one of the aforementioned categories.
  • the route of administration is selected from the group consisting of: oral, buccal, sublingual, transdermal, transmucosal, intranasal, intravenous ( .v.), intraperitoneal (i.p.), intramuscular (i.m.), subcutaneous (s.c.) or intrathecal (i.t).
  • buccal tablets or sublingual tablets may be used. These tablets are typically small, flat and soft, designed to be placed in the side of the cheek (i.e. buccal cavity) or designed to be placed under the tongue, to be directly absorbed through the buccal mucosa for a systemic effect.
  • Other dosage forms suitable for buccal administration are, for example, oral films administered on the gyngiva or tongue.
  • Sublingual spray is also a buccal formulation for delivery to the sublingual mucosa in the form of a spray for a systemic effect, typically provided in spray actuators, designed to access the mucosal surfaces under the tongue or the lips.
  • the composition may be provided in the form of a patch.
  • the major approaches for transdermal delivery include use of chemical penetration enhancers; physical enhancers, such as ultrasound, iontophoresis, electroporation, magnetophoresis, and microneedles; vesicles; particulate systems, such as those incorporating liposomes, niosomes, transfersomes, microemulsions, or solid lipid nanoparticles, as described for example in Rizwan et al, Recent Pat Drug Deliv Formul., 2009, 3(2): 105-24.
  • Suitable penetration enhancers for transderamal delivery include, for example, saturated and unsaturated fatty acids and their esters, alcohols, monoglycerides, diethanolamines, N,N- dimethylamines such as linolenic acid, linolenyl alcohol, oleic acid, oleyl alcohol, stearic acid, stearyl alcohol, palmitic acid, palmityl alcohol, myristic acid, myristyl alcohol, 1- dodecanol, 2-dodecanol, lauric acid, decanol, capric acid, octanol, caprylic acid, 1- dodecylazacycloheptan-2-one, ethyl caprylate, isopropyl myristate, hexamethylene lauramide, hexamethylene palmitate, capryl alcohol, decyl methyl sulfoxide, dimethyl sulfoxide, salicylic acid and its derivatives, N,N-diethyl-
  • Creams for transdermal delivery of flumazenil typically include gelling agents, for example, hydroxy methyl cellulose, hydroxypropyl cellulose, tragacanth, sodium alginate, gelatin, methylcellulose, sodium carboxymethylcellulose and polyvinyl alcohols.
  • gelling agents for example, hydroxy methyl cellulose, hydroxypropyl cellulose, tragacanth, sodium alginate, gelatin, methylcellulose, sodium carboxymethylcellulose and polyvinyl alcohols.
  • the efficacy of the methods described herein may be assessed, for example by direct observation of behavioral and physiological properties, by self-reporting, and/or by various well-known electrophysiological methods and performance skill methods.
  • Such methods include, for example, examining electroencephalograph (EEG) activity amplitude and frequency patterns, examining electromyogram activity, and examining the amount of time during a measurement time period, in which a mammal is awake or exhibits a behavioral or physiological property characteristic of wakefulness.
  • EEG electroencephalograph
  • Objective and subjective tests for wakefulness, alertness and performance include, for example, the Epworth Sleepiness Scale (Johns MW (1991) Sleep 14 (6): 540-5) and the Stanford Sleepiness Scale (Hoddes et al (1972) Psychophysiology 9: 150).
  • Additional methods used to monitor or assess alertness/drowsiness levels in a subject prior to and following use of the methods disclosed herein may employ various devices for measurement of eye position or closure, assumed to correlate with alertness/drowsiness, as disclosed for example in U.S. Patent Nos. 5,689,241; 5,682, 144 and 5,570,698.
  • flumazenil compositions according to the present invention may also be used in conjunction with the administration of other pharmaceutically active compounds.
  • the pharmaceutical composition according to the present invention may contain other pharmaceutically active compounds.
  • the methods of the invention further comprise co- administering, concomitantly or in sequence, the pharmaceutical composition comprising the flumazenil salt or flumazenil complex of the invention together with a wakefulness promoting agent.
  • the wakefulness promoting agent is selected from the group consisting of modafinil, armodafinil, adrafinil, methylphenidate, nefazodone, sodium oxybate, phentermine, pemoline, adrenaline, methylxantines, theobromine, caffeine and a combination thereof.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form described by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • Formulations with 1% flumazenil, in the form of a flumazenil salt or flumazenil complex, are presented in the following Tables. All formulations were evaluated for their appearance and only clear solutions, namely, clear solutions where a precipitate ('ppt') or slight precipitate ('sl-ppt') or a few particles ('part') are not formed, were tested for pH.
  • compositions comprising nicotinamide as a complexation agent and meglumine resulted in clear solutions at first. Twelve hours later, in formulations containing the meglumine a precipitate was formed. Formulation No. 2 containing 4.5% nicotinamide showed less precipitate (designated 'ppt' in the Tables) after 12 hours compared to formulation No. 1 containing 1.5% nicotinamide (Table 1). This result indicates that high nicotinamide concentrations may be preferred, although not necessary in order to achieve the concept of the present invention.
  • Nicotinamide/WFI (1/2 w/w) 1.5 4.5
  • Nicotinamide/WFI (1/2 w/w) 1.5 4.5
  • Example 2 Formulations of flumazenil (1.2 and 1.5 % w/w) nicotinamide complex
  • Formulations containing 1.2 to 1.5 % w/w flumazenil in the form of a flumazenil complex with nicotinamide were prepared and are presented in the following Table. Table 5. Flumazenil formulations with flumazenil nicotinamide complex (% w/w)
  • Formulations containing 1.2 % w/w flumazenil based on formulation 16 B were prepared with menthol and are presented in the following Table.
  • Niacinamide USP Natural Acids
  • C 6 H 6 N 2 0 122.12 g/mol
  • HPLC High-performance liquid chromatography
  • HPLC agents were HPLC grade water, methanol, acetonitrile, ammonium acetate, ammonium acetate and acetic acid glacial. Analysis was performed utilizing the column Inertsil, G.L. Sciences, ODS-3V, 5mm, 250x4.6 mm, C.N. 5020-01802 and the pre-column: Phenomex CI 8, 5 mm.
  • Formulation 16 (Table 4; 1% flumazenil), Formulation 16B (Table 5; 1.2% flumazenil) and Formulation 16B with menthol (Table 6; 1.2% flumazenil).
  • Solutions of flumazenil and of nicotinamide were made by mixing the dry compounds with the diluent (acetonitrile/water/acetic acid, 20/80/0.1 v/v/v).
  • Nicotinamide solution was injected and nicotinamide was eluted at retention time (RT) of 3.2 min. Good resolution was obtained between nicotinamide and flumazenil, since the retention time of flumazenil is about 9.4 min. Representative chromatograms are presented in Figures 2-4.
  • - Wstd is the weight of flumazenil standard in mg
  • - Dsmp is the dilution of the sample solution in ml (100 ml);
  • -Wsmp is the weight of sample solution in the sample in mg
  • Flumazenil chloride salt consisting of flumazenil and hydrochloride acid was prepared (Fig. 1).
  • the increased solubility of the flumazenil chloride salt enabled formulating a composition comprising flumazenil at a concentration of 0.66%.
  • the concentration of flumazenil in that composition is higher by about one order of magnitude than the concentration of flumazenil in the commercial product Romazicon® (Romazicon® contains 0.01% flumazenil).
  • the formulations were packed in 5-ml glass vials, capped with Teflon stoppers and crimped with aluminium seals. Vials were stored in a stability chamber at 25°C and 40°C. Stability was evaluated at the following stability time points: 2 weeks, 1 months and 3 months. At each stability time point samples were tested for: pH, appearance and flumazenil assay.
  • Toxicity of the flumazenil formulation of the invention is conducted in dogs. Four formulations are tested: vehicle (control), low dose, medium dose and higdose, according to the following study design:
  • Flumazenil is administered once a day, sublingually. The dose is sparyed under the tongue and is held there for at lest 5 minutes prior to swallowing.
  • mice All animals fast overnight (approximately 12 hours) prior to dosing and at least 2 hours postdose. Animals are observed twice daily and undergo detailed clinical observation weekly. The weekly observation include evaluating body weights and food consumption. Prior to initiation of the experiment, animals undergo physical examinations by staff veterinarian.
  • animal undergo electrocardiograms pretest, predose and postdose on day 1, predose and postdose prior to the terminal necropsy, and prior to the recovery necropsy.
  • Blood toxicokinetics is performed on blood samples collected on days 1 and 28 at six time points from each animal. Following necropsy, at the end of the experiment, weights of the follwing organs is measured: adrenals, brain, heart, kidneys, liver, lungs, ovaries with oviducts, pituitary, prostate, salivary glands, spleen, thyroid with parathyroid, thymus, testes and uterus. For all main study animals, a full set of standard tissues for slide preparation/microscopic pathology is collected (approximately 70). The target organs in recovery animals include gross lesions. A complete list of the target organs is given in Table 13 below. For toxicokinetic modeling standard parameters are evaluated, for example, AUC, ti/2, Tmax and Cmax. Data is analyzed by standard statistical analysis.
  • Lymph node (regional, if applicable) Trachea
  • inductive of sedative-hypnotic effects was induced by an intravenous injection of 20 mg/kg of benzodiazepine diazepam (Assival®) using a 24G sized needle over approximately 15 seconds. In case an animal was not sufficiently sedated an additional injection of 5 mg/kg was administrated. The day of induction is defined as "Day 1 ".
  • Test item (Formulation A or placebo) was administered via the sublingual route twice at an interval of approximately 2 minutes once the animal had no righting reflex about 5 minutes post the second injection of the diazepam.
  • Sleeping Time Determination was carried out as follows: upon diazepam injection the animal was placed on its back in a bedded standard cage. Sleeping time was determined when the test animal was no longer capable to maintain its Righting Reflex. This reflex is defined as the animal' s failure of righting 2 times within 30 seconds. Clock time was recorded after the first administration of the test item and again when the animal woke up. The sleeping time was determined as the elapsed time between first administration of the tested item and the time that the animal regained its Righting Reflex. The entire study was recorded (VCR).
  • the primary objective is to determine the single dose absolute bioavailability of sublingual (SL) Formulation A using the marketed IV flumazenil formulation as the comparator.
  • the secondary objectives are to characterize the concentration time course of two dose levels of SL Formulation A to support dose selection for Phase 2 and 3 studies and to evaluate the safety and tolerability of flumazenil formulations.
  • Study design includes masking and enrolment, open label, randomized, three-way crossover study, with 15 healthy subjects > 18 years of age, with no medication, based on the following study arms:
  • Study conditions standardized as much as possible in terms of die and fluid intake. This type of study is interventional.
  • the secondary outcome measure is to examine the safety and tolerability of Formulation A (l . lmg and 2.2mg), where the safety endpoints include: adverse event monitoring, vital signs, physical examinations, clinical laboratory tests and ECGs.
  • the time-points for blood and serum tests of flumazenil concentration are: 0, lOmin, 30min, 60min, 90min, 2h, 4h, 6h and 24h.
  • PK parameters for analysis are Cmax, Tmax, Cmin, Tmin, AUC0- ⁇ , AUC 0 -t and ⁇ ⁇ 2 .
  • the study is a Phase 2B Dose Range, Efficacy and Safety of sublingual Formulation A for the indication: reversal of the residual effects of GABA related (benzodiazepine (BNZ) and nonBNZ) hypnotic drugs in insomniac patients.
  • the residual sedative effect relates to benzodiazepine (BNZ) and nonBNZ hypnotic drugs.
  • the study evaluates different doses of Formulation A in comparison to a placebo.
  • Formulation A is provided as a sublingual (SL) spray formulation with flumazenil 1 lmg/ml.
  • Doses are determined with reference to the bioavailability study results.
  • GR Good Response
  • VGR Very Good Response to treatment
  • the secondary objectives are to evaluate the duration of action of a single dose of SL CRLS003 and to evaluate the safety of a single daily dose of Formulation A for 7 days.
  • Study Design is randomized, double blind, dose range and placebo controlled. The following examinations are carried out:
  • Each subject is in good health as determined by a medical history, physical examination and ECG.
  • Exclusion criteria are: 1. Using medications, except hypnotics and contraceptives, 1 month prior to screening.
  • CBT Cognitive Behavioral Therapy
  • the study includes the following visits:
  • Visit 1 A signed and dated informed consent form is collected from each participating subject, prior to any screening procedures. Screening visit takes place up to 14 days prior to treatment visit. Subjects who are screened are assigned a 3 -digit Screening number. During the screening visit, the following procedures are performed: Review Inclusion / Exclusion criteria; Medical history inquiry and demographic data; Record prior medication; Neurological assessment; Conduct physical examination including vital signs, height and weight measurements; Pregnancy test; 12-lead electrocardiography.
  • visit 3 is conducted applying same procedures as in visit plan 2.
  • Study Duration subjects participate in this study for 1 week of therapy and 1 week of follow-up. Interim analysis is carried (after 30 patients). Standard statistical methods are used to analyze the results (e.g. mean and standard deviation based on student-t test and ANOVA), where p ⁇ 0.05 defines significance.
  • This study is a double blind evaluation of SL Formulation A for reversal of next day residual sedative effect from hypnotic drugs used to treat insomnia. It is directed to evaluate the safety and effectiveness of SL Formulation A in insomniac patients in reversing the next day residual effect of hypnotic drugs.
  • the study is designed to collect short-term safety and tolerability data along with psycho motor/cognitive and behavioral function. It is an interventional type of study. The indication, drugs and treatment are similar to the those tested in the Phase 2B study.
  • Sample includes 300 subjects (150 per treatment group) randomized to flumazenil or placebo 1 : 1 ratio, stratified by BNZ or nonBNZ. Treatment response is evaluated for each treatment arm. Good Response (GR) is R*25%. Secondary objective include safety of SL Formulation A compared to placebo administered daily for 6 weeks.
  • the Study Design is randomized, double blind, placebo and controlled.
  • the purpose of this study is to evaluate mental score/quality of life improvement in patients with episodic FIE following the administration of sublingual flumazenil ( Formulation A).
  • This is an interventional type of study.
  • the indication is: relief of cognitive impairment symptoms in patients with episodic HE.
  • the investigational drug is Formulation A sublingual spray comprising 1.12mg /ml Flumazenil.
  • the time to recovery from episodic HE grade >2 is defined as a decrease of Conn score to Grade ⁇ 2, or a decrease in Asterixis.
  • the secondary objectives of this study are as follows:
  • SDMT Symbol digit modalities test
  • the study design is randomized, placebo controlled, double blinded administered sublingual flumazenil twice a day for 3 weeks. During study, patients with episodic HE grade >2 are screened for a respond to flumazenil therapy. Baseline assessment includes in addition to the routine testing described below: Clinical assessment (physical and blood tests), EEG, plasma assays for benzodiazepines and NCT.
  • the study population includes male and female, cirrhotic patients aged >18y, with episodic HE grade >2 .
  • Women of childbearing potential must have a negative pregnancy test at the screening visit and use a reliable method of contraception during the entire study duration (e.g. Contraceptive pill; Intra-uterine device; Contraceptive injection (prolonged-release gestagen); Subdermal implantation; Vaginal ring or Transdermal patch).
  • a reliable method of contraception e.g. Contraceptive pill; Intra-uterine device; Contraceptive injection (prolonged-release gestagen); Subdermal implantation; Vaginal ring or Transdermal patch).
  • Exclusion criteria include chronic BNZ therapy and epilepsy.
  • Safety assessment is based on changes from screening/baseline for clinical AEs reported by the subject, or observed by the investigator assessed by physical exam, oral and sublingual assessment, neurological assessment and vital signs.
  • Standard statistical methods are used to analyze the results (e.g. mean and standard deviation based on student-t test and ANOVA), where p ⁇ 0.05 defines significance.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Inorganic Chemistry (AREA)
  • Zoology (AREA)
  • Physiology (AREA)
  • Nutrition Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Neurology (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Biomedical Technology (AREA)
  • Toxicology (AREA)
  • Neurosurgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pyridine Compounds (AREA)

Abstract

The present invention provides soluble complexes of flumazenil, methods for the preparation thereof, pharmaceutical compositions comprising same and use of said compositions for alleviating or counteracting the various types of hypersomnia, drowsiness, residual effects associated with the administration of sleep/hypnotic drugs, alcohol intoxication or hepatic encephalopathy. The present invention is based in part on the unexpected discovery that flumazenil complexes are more soluble than flumazenil alone.

Description

FLUMAZENIL COMPLEXES, COMPOSITIONS COMPRISING SAME
AND USES THEREOF
FIELD OF THE INVENTION
The present invention provides soluble complexes of flumazenil, methods for the preparation thereof, pharmaceutical compositions comprising same and use of said compositions for alleviating or counteracting the various types of hypersomnia, drowsiness, residual effects associated with the administration of sleep/hypnotic drugs, sedation, alcohol intoxication and hepatic encephalopathy.
BACKGROUND OF THE INVENTION
Flumazenil (chemically named ethyl 8-fluoro- 5,6-dihydro- 5-methyl- 6-oxo- 4H- imidazo [1,5-a] [1,4] benzodiazepine- 3-carboxylate), also known as, Ro 15-1788, Anexate™, Lanexat™, Mazicon™ and Romazicon™, was initially disclosed in US 4,316,839. Flumazenil is an imidazobenzodiazepine having high affinity for the GAB AA/benzodiazepine-receptor complex, the specific binding site of benzodiazepines. As flumazenil is a competitive inhibitor of benzodiazepines, it is used for reversing benzodiazepine-induced sedation and anesthesia following therapeutic or diagnostic procedures (e.g. WO 2009/114740). Flumazenil is also known to reverse the effect of non- benzodiazepine drugs, such as the imidazopyridine hypnotic Zolpidem (e.g. Patat et al, Clin Pharmacol Then, 1994, 56(4):430-6). Flumazenil in combination with hydroxyzine and gabapentin are used for treating stimulant addiction (methamphetamine) (Bond AT, 1998, CNS Drugs, 9(1): 41-57). Flumazenil is also effective in treating hepatic encephalopathy (Als-Nielsen B et al. Cochrane Database of Systematic Reviews 2004, Issue 2. Art. No. : CD002798. DOI: 10.1002/14651858. CD002798.pub2). The aforementioned therapeutic effects were achieved by intravenous administration of flumazenil in a liquid formulation.
A liquid formulation of flumazenil (Romazicon®) is currently approved for reversing the sedative effects of benzodiazepines. Due to the limited solubility of flumazenil, the Romazicon® liquid formulation contains only 0.01% flumazenil. Poor water solubility frequently correlates with low drug absorption and bioavailability, and limits the amount of drug that can be administered in a pharmaceutical composition.
The use of hydrotopes, such as nicotinamide, for complexation with insoluble compounds in order to improve their solubility is known, for example, US 6,087,353.
There remains an unmet need to formulate flumazenil derivatives that are highly soluble and thereby suitable for the preparation of highly concentrated flumazenil formulations.
SUMMARY OF THE INVENTION
The present invention is directed to pharmaceutically acceptable complexes of flumazenil, comprising flumazenil and a complexation agent. The flumazenil complexes of the invention are highly soluble while maintaining the therapeutic activity known for flumazenil, namely, ameliorating or inhibiting any type of hypersomnia and drowsiness, including endogenous hypersomnia associated with GABA hyper-activation or exogenic hypersomnia e,g, residual hypersomnia associated with the administration of sleep/hypnotic drugs and ameliorating or inhibiting alcohol intoxication, or treating hepatic encephalopathy or any other disease or disorder associated with GABA related sedation or hypnotics. Having a significantly higher solubility of flumazenil, the flumazenil complexes of the invention offer a pronounced advantage over flumazenil compositions currently known in the art. Moreover, the flumazenil complexes of invention are advantageous for the preparation of formulations suitable for self-administration, such as, sublingual administration, thereby avoiding the involvement of professional care takers and the need of admitting to a health center.
The present invention is based in part on the unexpected discovery that flumazenil complexes are more soluble than flumazenil alone. Accordingly, these compounds are suitable for the preparation of highly concentrated pharmaceutical compositions of flumazenil. This enables administration of higher doses of flumazenil (per volume) than currently available. Typically, the concentration of flumazenil in the form of the flumazenil complexes of the invention is within the range of about 0.4 to 2%. This concentration is much higher than the concentration of flumazenil in commercially available formulations. In fact, the concentration of flumazenil in the pharmaceutical compositions of the invention is higher by more than two orders of magnitude from the concentration of flumazenil in the commercially available formulations. Highly concentrated flumazenil compositions are particularly advantageous in view of the maximal recommended daily dose approved for flumazenil solution (for example, Romazicon®), which is 3 mg. As the concentration of flumazenil in the commercial solution is only 0.01%, a dose of 3 mg requires administration of a large volume of the flumazenil solution, as high as 30 ml. The present invention overcomes this deficiency by providing concentrated formulations that can be administered in lower volumes thus improving patient compliance. Administering highly concentrated formulations of flumazenil also enables applying more routes of delivery, specifically, sublingual, submucosal, intramuscular, transdermal or any parentral route of administration.
Sublingual administration of the formulations of the invention is of special advantage. Apart from being easy to administer, the sublingual route bypasses the first liver metabolism, absorbance of flumazenil is immediate, thereby fast response to therapy can be achieved.
Although being highly concentrated, the formulations of the invention are stable and no precipitation occurs during storage. The stability of the formulations of the invention renders them suitable for long periods of storage.
According to a first aspect, the present invention provides a complex of ethyl 8-fluoro- 5,6-dihydro- 5-methyl- 6-oxo- 4H- imidazo [1,5-a] [1,4] benzodiazepine- 3-carboxylate (flumazenil) or a salt thereof and a complexation agent or derivatives thereof, with the proviso that the complexation agent is other than cyclodextrin or meglumine.
According to another aspect, the present invention provides a complex of ethyl 8- fluoro- 5,6-dihydro- 5-methyl- 6-oxo- 4H- imidazo [1,5-a] [1,4] benzodiazepine- 3- carboxylate (flumazenil) or a salt thereof and an aromatic stacking complexation agent.
According to another embodiment, the stacking complexation agent is selected from the group consisting of: benzoic acid, benzoate, hydroxybenzoic acid, hydroxybenzoate, salicylic acid, salicylate, nicotinamide, nicotinic acid, nicotinate, gentisic acid, gentisate, ethanolamide, toluic acid (ortho, meta or para), toluate (ortho, meta or para), aminobenzoic acid, aminobenzoate, anthranilic acid, anthranilate, butylmonoglycolsulfate and resorcinol.
According to yet another embodiment, the stacking complexation agent is selected from the group consisting of: benzoic acid, sodium benzoate, potassium benzoate, hydroxybenzoic acid, sodium hydroxybenzoate, potassium hydroxybenzoate, salicylic acid, sodium salicylate, potassium salicylate, nicotinamide, nicotinic acid, sodium nicotinate, potassium nicotinate, gentisic acid, sodium gentisate, potassium gentisate, ethanolamide, sodium toluate, toluic acid, potassium toluate, aminobenzoic acid, sodium aminobenzoate, potassium aminobenzoate, anthranilic acid, sodium anthranilate, potassium anthranilate, sodium butylmonoglycolsulfate, potassium butylmonoglycolsulfate and resorcinol.
According to yet another embodiment, the stacking complexation agent is selected from the group consisting of: sodium benzoate, sodium hydroxybenzoate, sodium salicylate, nicotinamide, sodium nicotinate, sodium gentisate, gentisic acid ethanolamide, sodium toluate, sodium aminobenzoates, sodium anthranilate, sodium butylmonoglycolsulfate and resorcinol.
It is noted that the complexation agent of the flumazenil complex of the current invention may be any complexation agent known in the art other than cyclodextrin. However, the present invention contemplates compositions comprising the flumazenil complexes of the invention, and cyclodextrin as an additional excipient.
According to one embodiment, the complexation agent is nicotinamide or a derivative thereof. According to yet another embodiment, the nicotinamide derivative is selected from the group consisting of: 2-amino-nicotinamide derivatives, 5 -phenyl-nicotinamide derivative and 6-substituted nicotinamide derivative.
According to yet another embodiment, the complexation agent: flumazenil ratio is in the range of 1 : 1 to 2: 1.
According to another aspect, the present invention provides a flumazenil nicotinamide stacking complex.
According to yet another aspect, the present invention provides a pharmaceutical composition comprising the flumazenil complex and a pharmaceutical acceptable carrier.
According to yet another embodiment, the pharmaceutical composition is in a form selected from the group consisting of: a pill, tablet, lozenge, coated tablet, granule, capsule, hard or soft gelatin capsule, aqueous solution, alcoholic solution, oily solution, syrup, emulsion suspension pastille suppository, solution for injection or infusion, ointment, tincture, cream, lotion, powder, spray, transdermal therapeutic systems, nasal spray, buccal spray, aerosol mixture, microcapsule, implant, rod and plaster. Each possibility represents a separate embodiment of the present invention.
According to yet another embodiment, the pharmaceutical composition is in a form selected from the group consisting of: immediate release, delayed release, pulsatile release, continuous release and repetitive release. Each possibility represents a separate embodiment of the present invention.
According to yet another embodiment, the composition is in a solid dosage form. According to yet another embodiment, the composition is suitable for oral administration.
According to yet another embodiment, flumazenil concentration in the pharmaceutical composition is within the range of about 0.4 to 2 % w/w. According to yet another embodiment, the concentration of flumazenil is within the range of about 0.5 to 1.8 % w/w. According to certain embodiments, the concentration of flumazenil is within the range of about 0.6 to 1.5 % w/w.
According to yet another embodiment, the pharmaceutical composition comprises flumazenil complex as an active ingredient, and a solubilizing agent selected from an alcohol, a glycol and a combination thereof. Each possibility represents a separate embodiment of the present invention.
According to yet another embodiment, the solubilizing agent comprises a combination of an alcohol and a glycol wherein the alcohol: glycol ratio is at least 1.5: 1. According to yet another embodiment, the alcohol: glycol ratio is in the range of 1.5: 1 to 5: 1.
According to yet another embodiment, the solubilizing agent comprises a combination of an alcohol and a glycol wherein the concentration of the solubilizing agent is at least 40%. According to yet another embodiment, the solubilizing agent comprises a combination of an alcohol and a glycol wherein the concentration of the solubilizing agent is in the range of 40% to 60%). According to yet another embodiment, the solubilizing agent comprises ethanol and propylene glycol.
According to yet another embodiment, the pharmaceutical composition further comprises a buffering agent.
According to yet another embodiment, the pharmaceutical composition further comprises at least one agent selected from the group consisting of: a penetration enhancer, a surfactant and cyclodextrin, as an inclusion complexing agent. Each possibility represents a separate embodiment of the present invention.
According to yet another embodiment, the cyclodextrin is hydroxypropyl β- cyclodextrin (HPCD). The cyclodextrin is preferably formulated in a buffer having a pH from about 3 to about 6. In one particular embodiment, the cyclodextrin (e.g., HPCD) is formulated in a citric acid buffer having a pH of about 4.
According to yet another embodiment, the preservative is selected from the group consisting of benzyl alcohol, propylparaben, methylparaben and combinations thereof. In one embodiment, the preservative is benzyl alcohol. In another embodiment, the preservative is a combination of propylparaben and methylparaben. Each possibility represents a separate embodiment of the present invention.
According to yet another embodiment, the penetration enhancer is menthol.
According to yet another embodiment, the buffering agent is selected from the group consisting of: citric buffer, sodium chloride and combination thereof. Each possibility represents a separate embodiment of the present invention.
According to yet another embodiment, the surfactant is a cationic surfactant. According to yet another embodiment, the surfactant is benzalkonium chloride.
According to yet another aspect, the present invention provides a method for treating a disease or disorder associated with binding of an agent to the GABA receptor, comprising administering to a patient in need thereof a pharmaceutical composition comprising as an active ingredient a flumazenil complex, and at least one pharmaceutical acceptable carrier.
According to one embodiment, treating a disease or disorder associated with binding of an agent to the GABA receptor comprises reversing the effect caused by the binding of the agent to the GABA receptor.
According to another embodiment, the agent is a benzodiazepine a non-benzodiazepine the binding of which to the GABA receptor causes sedation.
According to yet another aspect, the present invention provides a method for treating a disease or disorder comprising administering to a patient in need thereof a pharmaceutical composition comprising as an active ingredient a flumazenil complex, and at least one pharmaceutical acceptable carrier, wherein the disease or disorder is selected from the group consisting of: excessive sleepiness, alcohol intoxication and hepatic encephalopathy.
According to yet another aspect, the present invention provides a method for treating excessive sleepiness comprising administering to a patient in need thereof a pharmaceutical composition comprising as an active ingredient a flumazenil complex, and at least one pharmaceutical acceptable carrier.
According to one embodiment, the excessive sleepiness is selected from the group consisting of: excessive sleepiness associated with hypersomnia, excessive sleepiness associated with drowsiness, symptoms associated with overdose of a sleep drug, excessive sleepiness associated with sedation and anesthesia induced by a sleep drug, excessive sleepiness associated with sleep drug addiction, excessive sleepiness associated with stimulant addiction, Alzheimer's disease, anxiety, schizophrenia, specifically, schizophrenia associated with excessive sleepiness, the rebound effect of a sleep drug, balance impairment induced by a sleep drug or any combination thereof.
According to another embodiment, the excessive sleepiness is caused by alcohol intoxication.
According to yet another embodiment, the stimulant is methamphetamine. According to yet another embodiment, treating excessive sleepiness associated with stimulant addiction further comprises administering the flumazenil complex, or the pharmaceutical composition comprising same, in combination with hydroxyzine and gabapentin.
According to yet another embodiment, the sleep-drug is selected from the group consisting of a benzodiazepine, a benzodiazepine modulator, a benzodiazepine analog, a non- benzodiazepine, a 5-HT2A receptor antagonist, a melatonin receptor agonist, an orexin receptor antagonist, a selective serotonin reuptake inhibitor (SSRI), an antihistamine and an herbal product.
According to yet another embodiment, the sleep drug is a sedative/hypnotic drug.
According to yet another embodiment, the sleep drug is a benzodiazepine sedative/hypnotic drug. According to yet another embodiment, the benzodiazepine sleep drug is selected from the group consisting of alprazolam, bromazepam, clonazepam, clotiazepam, cloxazolam, diazepam, estazolam, etizolam, fludiazepam, flunitrazepam, flurazepam, halazepam, haloxazolam, lorazepam, medazepam, midazolam, nimetazepam, nitrazepam, olanzapine, oxazepam, quazepam, temazepam and triazolam.
According to yet another embodiment, treating excessive sleepiness comprises reversing benzodiazepine-induced sedation and anesthesia following therapeutic or diagnostic procedures.
According to yet another embodiment, the sleep-drug is a non-benzodiazepine sedative/hypnotic drug. According to yet another embodiment, the non-benzodiazepine sleep drug is selected from the group consisting of adipiplon (NG-2-73), agomelatine, almoxerant (ACT-078573), brotizolam, diphenhydramine, divaplon, doxepin, eplivanserin (SR 46349), doxylamine succinate, eszopiclone, indiplon, ocinaplon, pagoclone, pazinaclone, pruvanserin (EMD 281014), suproclone, suriclone, L-tryptophan, 5-hydroxy-L-tryptophan, melatonin, melatonin receptor agonists, such as VEC-162 and PD-6735, muramyl dipeptide, ramelteon, sleep-promoting substance, uridine, volinanserin (M- 100907), zaleplon, Zolpidem, imidazopyridine hypnotic Zolpidem, APD125, ACP-103, PD 200-390, HY10275, GW649863 and EVT-201.
According to yet another aspect, the present invention provides a method for treating alcohol intoxication comprising administering to a patient in need thereof a pharmaceutical composition comprising the flumazenil complex of the invention, and at least one pharmaceutical acceptable carrier.
According to one another embodiment, the excessive sleepiness caused by alcohol intoxication is treated by any one or more of reversing the effects of alcohol intoxication, reducing the effects of alcohol intoxication, alleviating the effects of alcohol intoxication and/or improving performance after alcohol consumption.
According to yet another aspect, the present invention provides a method for treating hepatic encephalopathy, comprising administering to a subject in need thereof the flumazenil complex of the invention or a pharmaceutical composition comprising same.
According to another embodiment, the route of administration is selected from the group consisting of: oral, buccal, sublingual, transdermal, transmucosal, intranasal, intravenous ( .v.), intraperitoneal (i.p.), intramuscular (i.m.), subcutaneous (s.c.) or intrathecal (i. t.). Each possibility represents a separate embodiment of the present invention.
According to yet another embodiment, the methods of the invention further comprise co-administering, concomitantly or in sequence, a pharmaceutical composition comprising a flumazenil complex together with a wakefulness promoting agent. According to yet another embodiment, the wakefulness promoting agent is selected from the group consisting of modafinil, armodafinil, adrafinil, methylphenidate, nefazodone, sodium oxybate, phentermine, pemoline, adrenaline, methylxantines, theobromine, caffeine and a combination thereof.
According to yet another aspect, the present invention provides a flumazenil complex for the treatment of excessive sleepiness or hepatic encephalopathy, the flumazenil complex comprises flumazenil and a complexation agent, with the proviso that the complexation agent is other than cyclodextrin or meglumine.
According to yet another aspect, the present invention provides a flumazenil complex for the treatment of excessive sleepiness or hepatic encephalopathy, the flumazenil complex comprises flumazenil and an aromatic complexation agent, such as, nicotinamide derivatives.
According to yet another aspect, the present invention provides a pharmaceutical composition comprising a flumazenil complex as the active ingredient, for the treatment of excessive sleepiness or hepatic encephalopathy.
Other objects, features and advantages of the present invention will become clear from the following description. BRIEF DESCRIPTION OF THE INVENTION
Figure 1 is a schematic presentation of a flumazenil hydrochloric acid salt.
Figure 2 exhibits a chromatogram of 0.6 mg/ml nicotinamide solution in diluents. Figure 3 shows a chromatogram of 40 μg/ml flumazenil solution in diluents. Figure 4 shows a chromatogram of a flumazenil nicotinamide complex in diluents. Figure 5 presents the body weight distribution of the rats (lF-treatment and 2F- placebo) in the study of reversal diazepam-induced sedative-hypnotic effects.
Figure 6 exhibits the sleeping time of the rats (lF-treatment and 2F-placebo) following treatment.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to pharmaceutically acceptable flumazenil salts comprising flumazenil and a counter-ion. In addition, the present invention provides pharmaceutically acceptable flumazenil complexes comprising flumazenil with a stacking complexation agent, such as an aromatic complexation agent, wherein the complexation agent is other than meglumine and cyclodextrin.
As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid salts of basic residues such as amines and the like. The pharmaceutically acceptable salts include conventional non-toxic salts of the parent compound formed, for example, from inorganic or organic acids, such as mineral acids (e.g., HC1) and nicotinic acids. Conventional non-toxic salts include those derived from inorganic acids, such as, hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids, such as, acetic, propionic, succinic, glycolic, stearic, lactic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
According to particular embodiments, the salts of the present invention are acid addition salts of flumazenil. According to some embodiments, the salts of the present invention are acid addition salts of flumazenil other than the acid addition salt flumazenil HC1.
Acid addition salts may, for example, be formed by mixing a solution of the compound of the present invention with a solution of a pharmaceutically acceptable acid, such as, hydrochloric acid, sulfuric acid, acetic acid, trifluoroacetic acid and benzoic acid.
The terms "acid salt" and "acid addition salts" are synonymous and refer to a class of salts formed by the partial neutralization of protic, diprotic or polyprotic acids. In diprotic or polyprotic acids, because the parent acid is only partially neutralized, one or more replaceable protons remain. Acid salt compounds can act either as an acid or a base: addition of a suitably strong acid will restore protons, and addition of a suitably strong base will remove protons. The pH of a solution of an acid salt will depend on the relevant equilibrium constants and the amounts of any additional base or acid.
According to some embodiments, the flumazenil complex of the invention comprises flumazenil and a complexation agent, wherein the complexation agent is preferably a hydrotope, other than cyclodextrins or meglumine, preferably, an aromatic stacking complexation agent.
The term "hydrotopes" as used herein, refers to compounds which are capable of opening up the water structure associated with hydrophobic (lipophilic) and other molecules. These compounds may be used to enhance the aqueous solubility of poorly water-soluble substances. Non limiting examples of hydrotopes includes sodium benzoate, sodium hydroxybenzoate, sodium salicylate, nicotinamide, sodium nicotinate, urea, sodium gentisate, gentisic acid ethanolamide, sodium toluate, sodium aminobenzoates, sodium anthranilate, sodium butylmonoglycolsulfate, resorcinol and the like. Each possibility represents a separate embodiment of the present invention.
As used herein, the term "stacking complexation agent" refers to complexation agent that forms a layered structure consisting of planar layers of the complexation agent stacked above and below the compounds with which the complex is formed. This mechanism is particularly suitable for complexing insoluble molecules having a relatively planar configuration, such as flumazenil. Preferably, the stacking complexation agent also has a relatively planar configuration. Preferred stacking complexation agents include nicotinamide and nicotinamide derivatives.
The term "nicotinamide derivatives" as used herein is meant to include any derivative of nicotinamide capable of forming a complex with flumazenil with improved solubility compared to the solubility of uncomplexed flumazenil. Acceptable nicotinamide derivatives include, but are not limited to, the nicotinamide derivatives disclosed in any one or more of: US Patent Nos. 7,056,934 and 7, 153,870; WO 2008/071665; Kuramochia et al, Bioorganic & Medicinal Chemistry, Vol. 13(12), 2005, 4022-4036; and Fonti et al, Nucl Med Biol, 1999, 26(6): 681-6, among others. Further non-limiting examples of nicotinamide derivatives include 2-amino-nicotinamide derivatives, 5 -phenyl-nicotinamide derivatives, and 6- substituted nicotinamide derivatives. Complex formation, which is non-covalent in nature, may be achieved by mixing appropriate ratios of the solvent-derived composition and the hydrotope or mixtures thereof in a suitable liquid vehicle, which may be aqueous, organic or a combination of both. Additional excipients, such as, surfactants, polyols, disaccharides may be added to facilitate complexation or for assisting in dispersability. The resultant complex may be isolated as a dry powder by any process known in the art (co-precipitation and drying, evaporation of the liquid vehicle, spray drying, lyophilization etc.). Particle size may be reduced by any standard technique known in the art, if desired. The resultant hydrotope complex may be used without further modification or may be compounded into a variety of other formulations or vehicles as required.
In a particular embodiment, the flumazenil stacking complex of the invention is a flumazenil nicotinamide complex. Nicotinamide, also known as niacinamide and nicotinic acid amide, is the amide of nicotinic acid (vitamin B3/niacin) and is also known as 3- pyridinecarboxamide, nicotinic acid amide and Vitamin PP.
Nicotinamide has over the past years been given at high doses for a variety of therapeutic applications. For regulatory purposes nicotinamide is classed as a food additive rather than a drug, a category that does require the formal safety evaluation normally expected of new drugs and therapies. Overall, and as detailed hereinafter, nicotinamide is considered safe. The therapeutic index of nicotinamide is wide but at very high doses (i.e. in megadoses) reversible hepatotoxicity has been reported in animals and humans. Minor abnormalities of liver enzymes can infrequently occur at the doses used for diabetes prevention. There is no evidence of teratogenicity from animal studies and there is no evidence of oncogenicity in human. Growth inhibition was shown in rats but growth in children is unaffected. In addition, minor degrees of insulin resistance attributed to nicotinamide have been reported. High-dose nicotinamide, i.e. in excess of 3 gm/day, should be considered as a drug with toxic potential at adult doses and unsupervised use should be discouraged.
The ratio of flumazenil: nicotinamide in the stacking complex of the invention is within the range of 1 : 1 to 1 :2. In certain embodiments the flumazenil: nicotinamide ratio in the complex of the invention is about 1 : 1.5. This ratio increases the solubility of flumazenil such that formulations comprising about 1% flumazenil with excellent solubility are obtained. The amount of nicotinamide in such formulations is about 1.5%. Accordingly, for example, a pharmaceutical composition comprising 3 mg of flumazenil, which is about the maximal daily dose allowed for flumazenil, corresponds to 4.5 mg of nicotinamide. An amount of 4.5 mg nicotinamide is lower by a factor of about 670 from 3 gr. (where 3 gr. is considered high dose and potentially toxic, for nicotinamide).
The marginal toxicity of nicotinamide renders it advantageous over other complexation agents. For example, cyclodextrins, another family of complexation agents, which increase the solubility of poorly soluble compounds by means of inclusion, are found toxic at relatively low concentrations, although there are significant differences between the cytotoxicity of the various cyclodextrin derivatives.
For example, the quantity of solubilized paclitaxel (as Taxol®), a poorly soluble drug, increased with cyclodextrin concentration. However, some of the cyclodextrins tested in this study were found toxic in mice with a maximum tolerated dose of 2 g/kg body weight, which is the quantity of cyclodextrin required to administer paclitaxel at 10 mg/kg. Other cyclodextrins allowed paclitaxel administration at higher doses yet had a maximum tolerated dose of 25 mg drug/kg.
Another advantage attributed to the flumazenil-nicotinamide complex of the invention is long shelf life. As exemplified hereinbelow, the flumazenil complex of the invention has an exceptional stability, during days, weeks and month of storage at room temperature and even at higher temperatures.
Without being bound to any theory or mechanism it is worth noting that cyclodextrins and nicotinamide increase solubility by different complexation mechanisms. It has been suggested that nicotinamide complex with hydrophobic ('guest') species is made out of a stack of nicotinamide and the guest species, whereby one or more 'sheets' of nicotinamide are stacked above and below a layer of the hydrophobic species. Clearly, stack formation depends on the 3D configuration of the members of the complex. While nicotinamide is substantially planar, for the formation of a stacked complex, it is generally required that the hydrophobic guest species would also be planar. Complex formation by cyclodextrins is exerted by way of inclusion. Cyclodextrins form a cavity, having an inner hydrophobic core and an outer hydrophilic core, where the cavity traps the hydrophobic species. The resulting complex exhibits modified physical and chemical properties, mostly in terms of water solubility. Cyclodextrins are relatively large molecules (molecular weight ranging from almost 1000 to over 1500), with a hydrated outer surface, and under normal conditions, cyclodextrin molecules will only permeate biological membranes with considerable difficulty. The present invention further provides a pharmaceutical composition comprising the flumazenil salt or complex as the pharmaceutically active ingredient and a pharmaceutical acceptable carrier.
The pharmaceutical compositions according to the present invention preferably comprise one or more pharmaceutical acceptable carriers and the active constituent(s). The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. The carriers in the pharmaceutical composition may comprise a binder, such as microcrystalline cellulose, polyvinylpyrrolidone (polyvidone or povidone), gum tragacanth, gelatine, starch, lactose or lactose monohydrate; a disintegrating agent, such as alginic acid, maize starch and the like; a lubricant or surfactant, such as magnesium stearate, or sodium lauryl sulphate; a glidant, such as colloidal silicon dioxide; a sweetening agent, such as sucrose or saccharin; and/or a flavoring agent, such as peppermint, methyl salicylate, or orange flavoring. More examples of the carriers include, but not limited to, physiological saline, polyethylene glycol, ethanol, vegetable oils, isopropyl myristate, etc. For topical administration, the compounds of the present invention can be formulated in the form of ointment or cream. Each possibility represents a separate embodiment of the present invention.
The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the composition and not deleterious to the recipients thereof.
The pharmaceutical composition comprising the flumazenil salt or complex of the invention as the active ingredient, may further comprise cyclodextrins, at least one solubilizing agent, at least one penetration enhancer, and at least one preservative. The pharmaceutical composition may further comprise additional excipients, such as, flavoring agents, among others.
The solubilizing agent is preferably a polar solvent such as mono- or poly-alcohols of linear or branched configuration (e.g., CI to C8 alcohols). Non-limiting examples include methanol, ethanol, propanol, iso-propanol, n-butanol, sec-butanol, isobutanol, t-butanol, n- pentanol, 2-pentanol, 3-pentanol, neopentanol, 1-hexanol, 2-hexanol, 3-hexanol, 1-heptanol, 2-heptanol, 1-octanol, as well as any geometrical isomers, enantiomers and diastereomers of any of the foregoing. Other suitable polar solvents include glycols such as ethylene glycol, propylene glycol and their polymers having a molecular weight between 400 and 1000. Each possibility represents a separate embodiment of the present invention. In one particular embodiment, the solubilizing agent is a combination of ethanol and propylene glycol. Preferred solubilizing agents include alcohols, glycols and combinations thereof. Each possibility represents a separate embodiment of the present invention.
According to some embodiments, the solubilizing agent in the composition of the invention comprises a combination of an alcohol and a glycol wherein the alcohol:glycol ratio is at least 1.5: 1 or in the range of 1.5: 1 to 5: 1. The solubilizing agent, preferably in the form of combination of an alcohol and a glycol, may be at least 40%, or in the range of 40% to 60%)., of the total weight of the pharmaceutical composition of the invention.
The expression "cyclodextrin" as used herein means α-, β- or γ- cyclodextrin or a derivative thereof. Suitable cyclodextrin derivatives for use in the formulations of the present invention include, but are not limited to the cyclodextrin listed above, e.g. hydroxypropyl derivatives of α-, β- and γ-cyclodextrin, sulfo alky 1 ether cyclodextrins such as sulfobutylether β-cyclodextrin, alkylated cyclodextrins such as the randomly methylated β-cyclodextrin, and various branched cyclodextrins such as glucosyl- and maltosyl β-cyclodextrin. Other cyclodextrins are described in US patent publication US 2004/0186075, the contents of which are incorporated by reference in their entirety.
In one currently preferred embodiment, the cyclodextrin is hydroxypropyl β- cyclodextrin (HPCD). The cyclodextrin is preferably formulated in a buffer having a pH from about 3 to about 6. In one particular embodiment, the cyclodextrin (e.g., HPCD) is formulated in a citric acid buffer having a pH of about 4.
The cyclodextrin component of the formulations of the present invention may be present in an amount from about 10%> to about 95% w/w, for example from about 30% to about 80%), from about 30% to about 75%, or about 60% based on the formulations of the invention. It is generally recognized that cyclodextrins act as true carriers by keeping the hydrophobic drug molecules in solution and delivering them to the surface of the biological membrane, e.g. skin, mucosa or the eye cornea, where they partition into the membrane. The relatively lipophilic membrane has low affinity for the hydrophilic cyclodextrin molecules and therefore they remain in the aqueous membrane exterior, e.g. the aqueous vehicle system, salvia or the tear fluid. Conventional penetration enhancers, such as alcohols and fatty acids, disrupt the lipid layers of the biological barrier. Cyclodextrins, on the other hand, may act as penetration enhancers by increasing drug availability at the surface of the biological barrier.
Therapeutic formulations suitable for oral administration, e.g. tablets and pills, may be obtained by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by mixing the constituent(s), and compressing this mixture in a suitable apparatus into tablets having a suitable size. Prior to the mixing, the phanquinone may be mixed with a binder, a lubricant, an inert diluent and/or a disintegrating agent and the further optionally present constituents may be mixed with a diluent, a lubricant and/or a surfactant. A tablet may be coated or uncoated. An uncoated tablet may be scored. A coated tablet may be coated with sugar, shellac, film or other enteric coating agents. Each possibility represents a separate embodiment of the present invention.
Therapeutic formulations suitable for parenteral administration include sterile solutions or suspensions of the active constituents. An aqueous or oily carrier may be used. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soy bean oil, mineral oil, sesame oil and the like. Formulations for parenteral administration also include a lyophilized powder comprising flumazenil and, optionally, further active constituents, that is to be reconstituted by dissolving in a pharmaceutically acceptable carrier that dissolves the active constituents, e.g. an aqueous solution of carboxymethylcellulose and lauryl sulphate. Each possibility represents a separate embodiment of the present invention.
When the pharmaceutical composition is a capsule, it may contain a liquid carrier, such as fatty oil, e.g. cacao butter.
Additional suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Each possibility represents a separate embodiment of the present invention.
The formulations of the present invention may further comprise penetration enhancers, such as menthol. Other penetration enhancers that may be used in the formulations of the present invention include, but are not limited to, anionic surfactants (e.g. sodium lauryl sulphate, sodium dodecyl sulphate), cationic surfactants (e.g. palmitoyl DL carnitine chloride, cetylpyridinium chloride), nonionic surfactants (e.g. polysorbate 80, polyoxyethylene 9-lauryl ether, glyceryl monolaurate, polyoxyalkylenes, polyoxyethylene 20 cetyl ether), lipids (e.g. oleic acid), bile salts (e.g. sodium glycocholate, sodium taurocholate), chitosan or a chitosan derivative, linalool, carvacrol, thymol, citral or t-anethole, and related compounds. Each possibility represents a separate embodiment of the present invention.
The formulations of the invention optionally further comprise at least one preservative. Any suitable preservative may be present in the formulation in the present invention. The preservative may be any pharmaceutically acceptable preservative, for example methyl 4- hydroxybenzoate (methyl paraben), ethyl 4-hydroxybenzoate (ethyl paraben), propyl 4- hydroxybenzoate (propylparaben), benzyl alcohol, sorbic acid, sodium benzoate, benzoic acid, and any combination thereof. Each possibility represents a separate embodiment of the present invention.
The formulation of the invention optionally further comprises a flavoring agent in an amount between 0.05 and 10 percent by weight of the total composition. In one embodiment, the flavoring agent is present in an amount between 0.1 and 2.5 percent by weight of the total composition. The flavoring agent is preferably selected from the group consisting of synthetic or natural oil of peppermint, oil of spearmint, citrus oil, fruit flavors, sweeteners (sugars, aspartame, saccharin, Estevia, etc.), and mixtures thereof. Menthol can also act as a flavoring agent.
In yet another embodiment, the therapeutic compound can be delivered in a controlled release system. The term "controlled release" is used herein to refer to a pharmaceutical dosage form in which release of the active ingredient is timed or modified to a rate sufficient to maintain the desired therapeutic level over an extended period of time. The release may be a "sustained release" or a "delayed release" such that release of the active ingredient from the pharmaceutical dosage form is other than promptly after administration of the dosage form, but rather is withheld or delayed following administration.
In one embodiment, a pump may be used. In another embodiment, polymeric materials can be used. In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, i.e., the central nervous system, thus requiring only a fraction of the systemic dose. Other controlled release systems are discussed, for example, in U.S. Patent No. 5, 120,548 which is directed a controlled-release drug delivery device comprised of swellable polymers. U.S. Patent No. 5,073,543 also describes controlled-release formulations containing a trophic factor entrapped by a ganglioside-liposome vehicle. U.S. Patent No. 5,639,476 discloses a stable solid controlled-release formulation having a coating derived from an aqueous dispersion of a hydrophobic acrylic polymer. Biodegradable microparticles are also known for use in controlled-release formulations. U.S. Patent No. 5,733,566 describes the use of polymeric microparticles that release antiparasitic compositions.
The controlled-release of the active ingredient may be stimulated by various inducers, for example, pH, temperature, enzymes, water, or other physiological conditions or compounds.
The terms "composition," "formulation" and "dosage form" are used herein interchangeably to encompass formulated preparations comprising one or more pharmacologically active drugs, and one or more pharmaceutically acceptable excipients, diluents or carriers. Compositions, formulations and dosage forms can be designed for administration by all possible administration routes to achieve the desired therapeutic response. The terms used may refer to the physical format of the product which is dispensed and administered to the patient, for example, a capsule or a patch. Alternately or in addition, the terms used may refer to any of: the mode of administration, the mode of delivery or the mode of release of the drug, for example a transdermal delayed release formulation.
The compounds of the present invention may be formulated into injections by dissolving, suspending or emulsifying water-soluble solvent such as saline and 5% dextrose, or in water-insoluble solvents such as vegetable oils, synthetic fatty acid glyceride, higher fatty acid esters and propylene glycol. The formulations of the invention may include any of conventional additives such as dissolving agents, isotonic agents, suspending agents, emulsifiers, stabilizers and preservatives.
According to yet another aspect, the present invention provides a method for treating a disease or disorder associated with binding of an agent to the GABA receptor, comprising administering to a patient in need thereof a pharmaceutical composition comprising an active ingredient selected from flumazenil salt and flumazenil complex, and at least one pharmaceutical acceptable carrier. Thus, the pharmaceutical composition of the invention is intended for use as an antidote for GABA related sedatives/hypnotics.
Preferably, the disease or disorder is excessive sleepiness, the agent is a benzodiazepine or a non-benzodiazepine, and the effect is sedation.
As used herein, the term "treating" encompasses substantially ameliorating, relieving, alleviating and preventing symptoms of a disease, disorder or condition in a subject.
As used herein, the term "administering" refers to delivery of a pharmaceutical compound to a subject by any means that does not affect the ability of the compound to perform its intended function.
The terms "sleep drug" is used herein in reference to pharmaceutical agents used for inducing and/or maintaining sleep, in particular, prescription sleep drugs that are classified as hypnotics/sedatives. The sleep-drug may be a sedative/hypnotic drug, a benzodiazepine, a benzodiazepine modulator, a benzodiazepine analog, a non-benzodiazepine, a 5-HT2A receptor antagonist, a melatonin receptor agonist, an orexin receptor antagonist, a selective serotonin reuptake inhibitor (SSRI), an antihistamine and an herbal product.
Suitable benzodiazepine sleep drugs include alprazolam, bromazepam, clonazepam, clotiazepam, cloxazolam, diazepam, estazolam, etizolam, fludiazepam, flunitrazepam, flurazepam, halazepam, haloxazolam, lorazepam, medazepam, midazolam, nimetazepam, nitrazepam, olanzapine, oxazepam, quazepam, temazepam and triazolam.
Suitable non-benzodiazepine sleep drug include adipiplon, agomelatine, almoxerant, brotizolam, diphenhydramine, divaplon, doxepin, eplivanserin, doxylamine succinate, eszopiclone, indiplon, ocinaplon, pagoclone, pazinaclone, pruvanserin (EMD 281014), suproclone, suriclone, L-tryptophan, 5-hydroxy-L-tryptophan, melatonin, melatonin receptor agonists, muramyl dipeptide, ramelteon, uridine, volinanserin, zaleplon, Zolpidem, imidazopyridine hypnotic Zolpidem, APD125, ACP-103, PD 200-390, HY10275, GW649863 and EVT-201
As used herein, the term "effective amount" refers to an amount of a pharmaceutical compound sufficient to achieve its desired effect.
According to one embodiment, the excessive sleepiness is selected from the group consisting of: excessive sleepiness associated with hypersomnia, excessive sleepiness associated with drowsiness, symptoms associated with overdose of a sleep drug, excessive sleepiness associated with alcohol intoxication, excessive sleepiness associated with sedation and anesthesia induced by a sleep drug, excessive sleepiness associated with sleep drug addiction, excessive sleepiness associated with stimulant addiction, the rebound effect of a sleep drug, balance impairment induced by a sleep drug or any combination thereof.
According to certain embodiments, treating excessive sleepiness comprises reversing benzodiazepine-induced sedation and anesthesia following therapeutic or diagnostic procedures.
As used herein, the term "hypersomnia" refers to chronic or recurrent bouts of excessive sleepiness, characterized by one or more of near-daily diurnal sleep episodes, excessive naps, abnormally prolonged sleep intervals, a perception of non-restorative sleep, and difficulty in making the transition from sleep to wakefulness. Hypersomnia may be one or more of: shift work sleep disorder; narcolepsy; obstructive sleep apnea/hypopnea syndrome; REM behavior disorder; frontal nocturnal dystonia; restless legs syndrome; nocturnal movement disorder; Kleine-Levin syndrome; Parkinson's disease; Alzheimer's disease, schizophrenia, anxiety, schizophrenia (in particular, schizophrenia associated with excessive sleepiness), excessive sleepiness; hypersomnia; idiopathic hypersomnia; recurrent hypersomnia; endozepine related recurrent stupor; and amphetamine resistant hypersomnia.
According to yet another embodiment, the excessive sleepiness is caused by alcohol intoxication.
According to yet another aspect, the method of the invention is directed to treating alcohol intoxication.
The term "alcohol intoxication" means overdose of alcohol (e.g., ethanol) leading to behavioral impairment. A person is said to suffer from alcohol intoxication when the quantity of alcohol the person consumes exceeds the individual's tolerance for alcohol and produces behavioral or physical abnormalities. In other words, the person's mental and physical abilities are impaired.
In this respect is it noted that the term alcohol refers to the generic term for ethanol, which is a particular type of alcohol produced by the fermentation of many foodstuffs— most commonly barley, hops, and grapes. Other types of alcohol commonly available such as methanol (common in glass cleaners), isopropyl alcohol (rubbing alcohol), and ethylene glycol (automobile antifreeze solution) are highly poisonous when swallowed, even in small quantities. Ethanol produces intoxication because of its depressive effects on various areas of the brain causing these impairments in a progressive order as the person gets more and more drunk. Symptoms of alcohol intoxication and/or impaired performance after alcohol consumption include disinhibition of normal social functioning (e.g., excessive talking), loss of memory, confusion, disorientation, uncoordinated movement, progressive lethargy, coma, or ultimately death.
According to one another embodiment, treating excessive sleepiness caused by alcohol intoxication is selected from the group consisting of: reversing the effects of alcohol intoxication, reducing the effects of alcohol intoxication, alleviating the effects of alcohol intoxication and improving performance after alcohol consumption.
According to yet another aspect, the method of the invention is directed treating hepatic encephalopathy with the flumazenil formulation.
The method of the invention provides an improved treatment of hepatic encephalopathy. First, the pharmaceutical compositions of the present invention are suitable for oral, sublingual or transdermal administration. Second, the pharmaceutical composition of the present invention includes much higher concentrations of flumazenil, of about 0.4 to 2%. This concentration is higher by two orders of magnitude from the concentration of flumazenil in the commercially available flumazenil formulations that are known to date. Thereby the pharmaceutical compositions and methods of the present invention provide advantageous patient compliance. In addition, the present invention provides attractive routes for the delivery of flumazenil (e.g. sub-mucosaland and transdermal) thereby averting the need for admission to health centers.
Without being bound by any theory or mechanism, administering the salts and complexes of the present invention by submucosal or sublingual or transdermal routes, bypasses the first pass effect (through the liver). This mechanism probably confers a material advantage in the treatment of the aforementioned diseases and disorders (residual effect of hypnotics, sedation, hepatic encephalopathy, etc.). Hepatic encephalopathy refers to a complex neuropsychiatric syndrome, which may complicate acute or chronic hepatic failure. It is characterized by changes in mental state including a wide range of neuropsychiatric symptoms ranging from minor not readily discernible signs of altered brain function, overt psychiatric and/or neurological symptoms to deep coma. Accordingly, the methods to estimate treatment effects and treatment outcomes are highly variable. The majority of hepatic encephalopathy occurs in patients with cirrhosis, often associated with spontaneous or iatrogenic portal-systemic shunting. Hepatic encephalopathy is generally considered a reversible metabolic encephalopathy. Traditionally, hepatic encephalopathy has been considered to be secondary to the accumulation of toxic products, which have not been metabolized by the liver. Various hypotheses have been suggested, e.g., alterations in the permeability of the blood-brain barrier, abnormal neurotransmitter balance, altered cerebral metabolism, and increased amounts of endogenous benzodiazepine-like compounds - the gamma-amino butyric acid (GABA)/benzodiazepine hypothesis. GABA is the principal inhibitory neurotransmitter in mammals that acts by binding to a receptor on a 'supramolecular complex' called the GABA/benzodiazepine complex, which also has binding sites for benzodiazepines and barbiturates. By binding to the GABA/benzodiazepine complex, benzodiazepines cause sedation through neural inhibition. It has been suggested that liver failure leads to the accumulation of substances that bind to the GABA/benzodiazepine complex resulting in neural inhibition which may progress to coma. Accordingly, a benzodiazepine-receptor antagonist, flumazenil, has been assessed in the treatment of hepatic encephalopathy in the hope of reversing neuropsychiatric symptoms related to the accumulation of endogenous benzodiazepine.
As detailed above, Als-Nielsen et al. (ibid) disclose the use of flumazenil, administered via intravenous infusion of 1 mg flumazenil in 20 ml saline solution (0.005%) over 3-5 minutes, for treating hepatic encephalopathy. Als-Nielsen reports that flumazenil had a significant beneficial effect on short-term improvement of hepatic encephalopathy in patients with cirrhosis and a highly favorable prognosis but had no significant effect on recovery or survival.
The methods of the invention may be used in conjunction with insomnia treatment modalities, and serve to eliminate or diminish residual soporific effects associated with administration of sleep drugs. The invention is effective for counteracting excessive sleepiness induced by a wide variety of sleep drugs. Such sleep drugs include benzodiazepine and non-benzodiazepine drugs which are classified as hypnotics/sedatives, as well as other prescription and non-prescription sleep drugs, including those classified as 5-HT2A receptor antagonists, melatonin receptor agonists, orexin receptor antagonists, selective serotonin reuptake inhibitors (SSRIs), and other sleep inducing agents such as antihistamines, melatonin and certain herbal products. It is to be specifically understood that a particular sleep drug may be classified under more than one of the aforementioned categories.
According to another embodiment, the route of administration is selected from the group consisting of: oral, buccal, sublingual, transdermal, transmucosal, intranasal, intravenous ( .v.), intraperitoneal (i.p.), intramuscular (i.m.), subcutaneous (s.c.) or intrathecal (i.t).
For buccal administration, buccal tablets or sublingual tablets may be used. These tablets are typically small, flat and soft, designed to be placed in the side of the cheek (i.e. buccal cavity) or designed to be placed under the tongue, to be directly absorbed through the buccal mucosa for a systemic effect. Other dosage forms suitable for buccal administration are, for example, oral films administered on the gyngiva or tongue.
Sublingual spray is also a buccal formulation for delivery to the sublingual mucosa in the form of a spray for a systemic effect, typically provided in spray actuators, designed to access the mucosal surfaces under the tongue or the lips.
For transdermal delivery of the composition of the invention, the composition may be provided in the form of a patch. The major approaches for transdermal delivery include use of chemical penetration enhancers; physical enhancers, such as ultrasound, iontophoresis, electroporation, magnetophoresis, and microneedles; vesicles; particulate systems, such as those incorporating liposomes, niosomes, transfersomes, microemulsions, or solid lipid nanoparticles, as described for example in Rizwan et al, Recent Pat Drug Deliv Formul., 2009, 3(2): 105-24.
Suitable penetration enhancers for transderamal delivery include, for example, saturated and unsaturated fatty acids and their esters, alcohols, monoglycerides, diethanolamines, N,N- dimethylamines such as linolenic acid, linolenyl alcohol, oleic acid, oleyl alcohol, stearic acid, stearyl alcohol, palmitic acid, palmityl alcohol, myristic acid, myristyl alcohol, 1- dodecanol, 2-dodecanol, lauric acid, decanol, capric acid, octanol, caprylic acid, 1- dodecylazacycloheptan-2-one, ethyl caprylate, isopropyl myristate, hexamethylene lauramide, hexamethylene palmitate, capryl alcohol, decyl methyl sulfoxide, dimethyl sulfoxide, salicylic acid and its derivatives, N,N-diethyl-m-toluamide, 1 -substituted azacycloalkan-2-ones, propylene glycol, polyethylene and glycol monolaurate. Any compound compatible with flumazenil, and that has transdermal permeation enhancing activity may be selected.
Creams for transdermal delivery of flumazenil typically include gelling agents, for example, hydroxy methyl cellulose, hydroxypropyl cellulose, tragacanth, sodium alginate, gelatin, methylcellulose, sodium carboxymethylcellulose and polyvinyl alcohols.
The efficacy of the methods described herein may be assessed, for example by direct observation of behavioral and physiological properties, by self-reporting, and/or by various well-known electrophysiological methods and performance skill methods. Such methods include, for example, examining electroencephalograph (EEG) activity amplitude and frequency patterns, examining electromyogram activity, and examining the amount of time during a measurement time period, in which a mammal is awake or exhibits a behavioral or physiological property characteristic of wakefulness.
Objective and subjective tests for wakefulness, alertness and performance include, for example, the Epworth Sleepiness Scale (Johns MW (1991) Sleep 14 (6): 540-5) and the Stanford Sleepiness Scale (Hoddes et al (1972) Psychophysiology 9: 150).
Additional methods used to monitor or assess alertness/drowsiness levels in a subject prior to and following use of the methods disclosed herein may employ various devices for measurement of eye position or closure, assumed to correlate with alertness/drowsiness, as disclosed for example in U.S. Patent Nos. 5,689,241; 5,682, 144 and 5,570,698.
The flumazenil compositions according to the present invention may also be used in conjunction with the administration of other pharmaceutically active compounds. In addition, the pharmaceutical composition according to the present invention may contain other pharmaceutically active compounds.
According to some embodiments, the methods of the invention further comprise co- administering, concomitantly or in sequence, the pharmaceutical composition comprising the flumazenil salt or flumazenil complex of the invention together with a wakefulness promoting agent. According to some embodiments, the wakefulness promoting agent is selected from the group consisting of modafinil, armodafinil, adrafinil, methylphenidate, nefazodone, sodium oxybate, phentermine, pemoline, adrenaline, methylxantines, theobromine, caffeine and a combination thereof.
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such container(s) can be a notice in the form described by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
The following examples are presented in order to more fully illustrate certain embodiments of the invention. They should in no way, however, be construed as limiting the broad scope of the invention. One skilled in the art can readily devise many variations and modifications of the principles disclosed herein without departing from the scope of the invention.
EXAMPLES
Example 1. Formulations of flumazenil salts and complexes at 1% flumazenil
Formulations with 1% flumazenil, in the form of a flumazenil salt or flumazenil complex, are presented in the following Tables. All formulations were evaluated for their appearance and only clear solutions, namely, clear solutions where a precipitate ('ppt') or slight precipitate ('sl-ppt') or a few particles ('part') are not formed, were tested for pH.
Compositions comprising nicotinamide as a complexation agent and meglumine resulted in clear solutions at first. Twelve hours later, in formulations containing the meglumine a precipitate was formed. Formulation No. 2 containing 4.5% nicotinamide showed less precipitate (designated 'ppt' in the Tables) after 12 hours compared to formulation No. 1 containing 1.5% nicotinamide (Table 1). This result indicates that high nicotinamide concentrations may be preferred, although not necessary in order to achieve the concept of the present invention. The appropriate pH values of a formulation intended for sublingual administration are between 4.0 to 9.0. The pH values of the nicotinamide- containing formulations meet this requirement (pH = 5.0±0.2).
Table 1. Formulations of flumazenil-nicotinamide and flumazenil meglumine (% w/w)
Material name 1 2 3 4 5 6
Flumazenil 1 1 1 1 1 1
Ethanol 25 25 25
Propylene glycol 25 25 25
Citric buffer lOmM pH 4.0 47.4 44.4 48.7
Menthol in EtOH (1/1 w/w) 0.1 0.1 0.1
Nicotinamide/WFI (1/2 w/w) 1.5 4.5
Benzalkonium chloride
0.2
(BKC) 50% aq. solution
Water (WFI) 98.67 98.67 98.356
HC1 0.33
H2S04 0.33
Meglumine 0.644
Appearance t=0 clear clear ppt ppt ppt ppt
Appearance T=12h sl-ppt sl-ppt
pH 5.06 5.15
Table 2. Formulations with cyclodextrin (FIPCD; % w/w)
Material name 1' V 3' 4' 5' 6'
Flumazenil 1 1 1 1 1 1
Ethanol 20 20 20
Propylene glycol 20 20 20
30% HPCD in citric buffer 57.4 54.4 58.7
30% HPCD in water 98.67 98.67 98.36
Menthol in EtOH (1/1 w/w) 0.1 0.1 0.1
Nicotinamide/WFI (1/2 w/w) 1.5 4.5
BKC 50% aq. solution 0.2
HC1 0.33
H2S04 0.33
Meglumine 0.644
Appearance t=0 ppt ppt ppt ppt ppt ppt
Table 3. Formulations with flumazenil meglumine complex (% w/w)
Material name 7 8 9 8' 9'
Flumazenil 1 1 1 1 1
Ethanol 25 25 25 20 20
Propylene glycol 25 25 25 20 20
WFI 47.4 44.4
30% HPCD in water 57.4 54.4
Citrate buffer 44.4
Menthol in EtOH (1/1 w/w) 0. 1 0. 1 0. 1 0.1 0. 1
Meglumine in WFI (1/2 w/w) 1.5 4.5 4.5 1.5 4.5
Appearance t=0 clear clear clear ppt ppt
Appearance t=12 h si- ppt si- ppt ppt
pH 10.91 1 1.32 10.50
Table 4. Formulations with flumazenil nicotinamide complex and flumazenil meglumine complex (% w/w)
Material name 10 11 12 13 14 15 16
Flumazenil 1 1 1 1 1 1 1
Ethanol 25 25 25 30 35 35 40
Propylene glycol 25 25 25 20 20 15 10
Citric buffer lOmM pH 4.0 41.5 46.9 43.9 44.5 39.5 44.5 44.5
Nicotinamide/WFI (1/2 w/w) 7.5 1.5 4.5 4.5 4.5 4.5 4.5
Meglumine in WFI (1/2 w/w) 0.6 0.6
Appearance t=0 clear clear clear clear clear clear clear pH, T O 5. 10 9.02 9.12 5.09 5.25 5.16 5. 17
Appearance t=24-72h sl-ppt ppt ppt sl-ppt clear clear clear
Appearance t=24-72h 10.91 1 1.32 10.50
Example 2. Formulations of flumazenil (1.2 and 1.5 % w/w) nicotinamide complex
Formulations containing 1.2 to 1.5 % w/w flumazenil in the form of a flumazenil complex with nicotinamide were prepared and are presented in the following Table. Table 5. Flumazenil formulations with flumazenil nicotinamide complex (% w/w)
Material name 14A 15A 16A 14B 16B 17 18 19 20
Flumazenil 1.5 1.5 1.5 1.2 1.2 1.5 1.5 1.5 1.5
Ethanol 35 35 40 35 40 40 35 40 30
Propylene glycol 20 15 10 20 10 20 20 10 30
Citric buffer
39 44 44 39.3 44.3 34 36 41 34 10mM pH 4.0
Nicotinamide/WFI
4.5 4.5 4.5 4.5 4.5 4.5 7.5 7.5 4.5 (1/2 w/w)
Appearance T-0 clear clear clear clear clear clear clear clear clear pH, T-0 5.27 5.18 5.24 5.25 5.26 5.44 5.28 5.26 5.30
Appearance T-24-72h sl-ppt ppt sl-ppt clear clear clear ppt ppt ppt
Appearance T-lweek part clear part
Example 3. Formulations of flumazenil nicotinamide complexes and menthol
Formulations containing 1.2 % w/w flumazenil based on formulation 16 B (Table 6, above), were prepared with menthol and are presented in the following Table.
Table 6. Flumazenil formulations with 1.2 flumazenil (% w/w) and menthol
0.2%menthol 0.1%menthol
Material name
% g % g
Flumazenil 1.2 0.06 1.2 0.06
Ethanol 40 2 40 2
Propylene glycol 10 0.5 10 0.5
Citric buffer lOmM pH 4.0 43.9 2.195 44.1 2.205
Nicotinamide/WFI (1/2 w/w) 4.5 0.225 4.5 0.225
Menthol/EtOH (1/1 w/w) 0.4 0.02 0.2 0.01
Appearance T-0 clear clear
pH, T-0 5.17 5.25
Appearance T-24h part clear
The results are similar to the results obtained in the absence of menthol (see Tables 4-5, above) indicating that menthol is not necessary in order to achieve the concept of the present invention, particularly, the improved solubility of the flumazenil salts or complexes. Example 4. HPLC analysis of flumazenil in the presence of nicotinamide
The conditions for determining by HPLC the presence of flumazenil in solutions containing flumazenil complex (1% (10 mg/g) or 1.2% (12 mg/g) flumazenil and 1.5% nicotinamide), were determined.
The details of the tested compounds were as follows:
1. Flumazenil [C15H14FN3O3], 303.29 g/mol (Chemagis, purity: 99.3%,)
2. Niacinamide USP (Nicotinamide) [C6H6N20], 122.12 g/mol (Spectrum, purity:
99.5%)
The HPLC (High-performance liquid chromatography) agents were HPLC grade water, methanol, acetonitrile, ammonium acetate, ammonium acetate and acetic acid glacial. Analysis was performed utilizing the column Inertsil, G.L. Sciences, ODS-3V, 5mm, 250x4.6 mm, C.N. 5020-01802 and the pre-column: Phenomex CI 8, 5 mm.
For Mobile Phase A, 13 mM Ammonium acetate buffer in water (pH adjusted at 4.40 with acetic acid) was utilized and acetonitrile for Mobile Phase B. Analysis was performed at column temperature of 30° ± 5°C, autosampler temperature of 10° ± 5°C, injection volume of 20 ml, PDA programmed 210 to 330 nm, sampling rate = 2, resolution = 1.2 and monitoring wavelength: 247 nm. The resulting gradient Table is given below.
Table 7. HPLC calibration
Time (min) Flow (ml/min) %A %B Curve
0 1 80 20 6
0.5 1 80 20 6
8.0 1 50 50 6
8.01 1.2 50 50 6
13.0 1.2 50 50 6
13.01 1.2 80 20 6
19.0 1 80 20 6
Samples of the following liquid formulations were prepared: Formulation 16 (Table 4; 1% flumazenil), Formulation 16B (Table 5; 1.2% flumazenil) and Formulation 16B with menthol (Table 6; 1.2% flumazenil). Solutions of flumazenil and of nicotinamide were made by mixing the dry compounds with the diluent (acetonitrile/water/acetic acid, 20/80/0.1 v/v/v).
Nicotinamide solution was injected and nicotinamide was eluted at retention time (RT) of 3.2 min. Good resolution was obtained between nicotinamide and flumazenil, since the retention time of flumazenil is about 9.4 min. Representative chromatograms are presented in Figures 2-4.
Assay of flumazenil solutions was determined using single point calibration. Flumazenil standard solution 40ug/ml was injected five times. Flumazenil sample solutions were prepared in triplicate and injected once. The assay (%) of flumazenil in a sample was calculated according to the following formula:
Assay (%) = (Wstd x P x Dsmp x Asmp/(Dstd x Astd x Dose x Wsmp)) xlOO Where:
- Wstd is the weight of flumazenil standard in mg;
- P is the purity of flumazenil standard (99.3%);
- Dstd is the dilution factor of the calibration solution (500);
- Dsmp is the dilution of the sample solution in ml (100 ml);
-Wsmp is the weight of sample solution in the sample in mg;
- Dose is the theoretical amount (%) of flumazenil in the solution;
- Astd is an average peak area of flumazenil for the five first injections of flumazenil calibration solution;
- Asmp is an average peak area of Flumazenil in the sample.
The assay results are summarized in the Table below.
Table 8. Flumazenil formulation analysis
Formulation Sample weight Peak area Assay (%) Mean assay (%)
No. (mg) (%)
16 412.18 2956920 103.64 103.8
411.98 2964443 103.96
16B 338.43 2744305 97.63 100.9
337.90 2863082 102.01
339.55 2907803 103.1
16B with 327.52 2803639 103.06 101.6
menthol 328.91 2757927 100.95
332.91 2783892 100.68
The results indicate that under the tested conditions there is no interference of nicotinamide signal with the signal of flumazenil. Accordingly, the aforementioned FIPLC parameters are appropriate for evaluating the content of flumazenil in formulations comprising the flumazenil nicotinamide complex of the invention. Example 5. HC1 acid addition salt of flumazenil
Flumazenil chloride salt consisting of flumazenil and hydrochloride acid was prepared (Fig. 1). The increased solubility of the flumazenil chloride salt enabled formulating a composition comprising flumazenil at a concentration of 0.66%. The concentration of flumazenil in that composition is higher by about one order of magnitude than the concentration of flumazenil in the commercial product Romazicon® (Romazicon® contains 0.01% flumazenil).
Example 6. Stability analysis
The short term stability (STS) of flumzenil-nicotinamide formulations was tested on two flumazenil solutions (Table 9).
Table 9. Formulations participated in the STS study (% w/w)
Material Batch No. 1 Batch No. 2
Flumazenil 1 1.2
Ethanol (absolute) 40.1 40.1
Propylene glycol 10 10
Citric acidic anhydrous 0.05 0.05
Niacinamide (nicotinamide) 1.5 1.5
Menthol 0.1 0.1
Water for injection 47.2 47.0
The formulations were packed in 5-ml glass vials, capped with Teflon stoppers and crimped with aluminium seals. Vials were stored in a stability chamber at 25°C and 40°C. Stability was evaluated at the following stability time points: 2 weeks, 1 months and 3 months. At each stability time point samples were tested for: pH, appearance and flumazenil assay.
The results are summarized in Table 10 below. The assay values for flumazenil were in the rangeg of 90-110% and did not change over 3-months storge at 25°C and at 40°C. The pH of the formulations remained in the range of 5.13-5.39 throughout the experiment (3 months) under all temperatures. Furthermore, the solutions remained clear during storage in the stability chambers over the period and storage conditions that were tested. Thus, the results clearly indicate that the flumazenil-niacinamide formulations are stable and suitable for long shelf lives. Table 10. STS study
Time point Batch No. 1 2
API content 1% 1.2%
Storage 25°C 40°C 25°C 40°C
Temperature
T-0 pH 5.17 5.20
Assay (%) 105.1 107.9
Appearance clear clear clear clear
T-2 weeks pH 5.24 5.26 5.30 5.34
Assay (%) 100.6 102.2 100.3 102.2
Appearance clear clear clear clear
T-l month pH 5.24 5.38 5.31 5.39
Assay (%) 99.5 100.2 99.6 98.4
Appearance clear clear clear clear
T-3 months pH 5.13 5.13 5.28 5.25
Assay (%) 103.0 102.7 102.9 103.4
Appearance clear clear clear clear
Example 7. Toxicity studies in doss
Toxicity of the flumazenil formulation of the invention is conducted in dogs. Four formulations are tested: vehicle (control), low dose, medium dose and higdose, according to the following study design:
Table 11. Toxicity study design
Main Study Recovery
Dose Male Female Male Female
Vehicle 3 3 2 2
Low dose 3 3 2 2
Medium dose 3 3 2 2
High dose 3 3 2 2
Flumazenil is administered once a day, sublingually. The dose is sparyed under the tongue and is held there for at lest 5 minutes prior to swallowing.
All animals fast overnight (approximately 12 hours) prior to dosing and at least 2 hours postdose. Animals are observed twice daily and undergo detailed clinical observation weekly. The weekly observation include evaluating body weights and food consumption. Prior to initiation of the experiment, animals undergo physical examinations by staff veterinarian.
All animals pretest and at termination and recovery undergo ophthalmology evaluation.
In addition, animal undergo electrocardiograms pretest, predose and postdose on day 1, predose and postdose prior to the terminal necropsy, and prior to the recovery necropsy.
Clinical pathology (hematology, coagulation, clinical chemistry and urinalysis) is obtained for animals pretest, and all survivors prior to the terminal and recovery necropsies. The tests are listed in Table 12 below.
Blood toxicokinetics is performed on blood samples collected on days 1 and 28 at six time points from each animal. Following necropsy, at the end of the experiment, weights of the follwing organs is measured: adrenals, brain, heart, kidneys, liver, lungs, ovaries with oviducts, pituitary, prostate, salivary glands, spleen, thyroid with parathyroid, thymus, testes and uterus. For all main study animals, a full set of standard tissues for slide preparation/microscopic pathology is collected (approximately 70). The target organs in recovery animals include gross lesions. A complete list of the target organs is given in Table 13 below. For toxicokinetic modeling standard parameters are evaluated, for example, AUC, ti/2, Tmax and Cmax. Data is analyzed by standard statistical analysis.
Table 12: Standard clinical pathology tests
Standard hematology parameters evaluated
Absolute and percent reticulocytes Mean corpuscular hemoglobin
Erythrocyte count Mean corpuscular hemoglobin concentration
Hematocrit Mean corpuscular volume
Hemoglobin Platelet count
Leukocyte count (total and differential) Reticulocytes
Standard coagulation parameters evaluated
Prothrombin time Activated partial thromboplastin time
Standard clinical chemistry parameters evaluated
Albumin/Globulin Ratio (calculated) Glucose
Alanine aminotransferase Phosphorus
Albumin Potassium
Alkaline phosphatase Sodium
Aspartate aminotransferase Sorbitol dehydrogenase
Calcium Total bilirubin (with direct if total exceeds 1 mg/dL)
Chloride Total cholesterol
Creatinine Total protein
Gamma glutamyl transferase Triglycerides
Globulin (calculated) Urea nitrogen
Standard urinalysis parameters evaluated
Bilirubin pH
Color and appearance Protein
Glucose Specific gravity
Ketones Total volume
Microscopy of spun deposit Urobilinogen
Occult blood Table 13: Standard microscopic tissue list
Standard microscopic pathology for dogs
Adrenal gland Pancreases
Aorta Pituitary
Bone with bone marrow, femur Parathyroid gland
Bone with bone marrow, sternum Prostate
Bone with bone marrow, rib Salivary gland, mandibular
Bone marrow smear Salivary gland, parotid
Brain Salivary gland, sublingual
Epididymis Skeletal muscle, biceps femoris
Esophagus Skin
Eye (with optic nerve) Small intestine, duodenum
Gallbladder Small intestine, ileum with Peyers
Heart Small intestine, jejunum
Joint, tibiofemoral Spinal cord, cervical
Kidney Spinal cord, lumbar
Large intestine, cecum Spinal cord, thoracic
Large intestine, colon Spleen
Large intestine, rectum Stomach, cardia
Larynx Stomach, fundus
Liver Stomach, pylorus
Lung Testis
Lymph node (mandibular) Thymus
Lymph node (mesenteric) Thyroid gland
Lymph node (tracheobronchial) Tongue
Lymph node (regional, if applicable) Trachea
Mammary gland (process females only) Urinary bladder
Nerve (sciatic) Uterus with cervix
Nictitans gland Vagina
Ovary Gross lesions
Oviducts Tissue masses
Example 8. Reversal of diazepam-induced sedative-hypnotic effects in rats
The effect of flumazenil nicotinamide formulation A (Table 15) on reversal of diazepam induced sedative- hypnotic state was studied in SD rat model. The study end point was set by determining the sleeping time of the rats (females, 8-9 weeks of age, n=8) in each group. The experiment was conducted in two groups, of four rats each:
Table 14. Treatment groups
Group No. N= Treatment
IF 4 Flumazenil
Formulation A
2F 4 Placebo (vehicle)
Following acclimation (5 days) inductive of sedative-hypnotic effects was induced by an intravenous injection of 20 mg/kg of benzodiazepine diazepam (Assival®) using a 24G sized needle over approximately 15 seconds. In case an animal was not sufficiently sedated an additional injection of 5 mg/kg was administrated. The day of induction is defined as "Day 1 ".
The test item (Formulation A or placebo) was administered via the sublingual route twice at an interval of approximately 2 minutes once the animal had no righting reflex about 5 minutes post the second injection of the diazepam.
The components of the placebo solution are listed in Table 15.
Table 15. Placebo and flumazenil solutions for sublingual administration in rats.
Component Formulation A Placebo
% w/w
Flumazenil 1.1 -
Ethanol 40 10
Propylene glycol 10 20
30% HPCD in citric buffer 59.3
10mM pH 4.0
Menthol in EtOH (1/1 w/w) - 0.1
Propylparaben/Methylparaben - 10.2
in EtOH 0.02/0.18/10 w/w/w
Citric acid anyhdrous 0.05 -
Sodium citrate dehydrate 0.05 -
Nicotinamide 1.5 -
L-menthol 0.1 -
Water 47 -
Sleeping Time Determination was carried out as follows: upon diazepam injection the animal was placed on its back in a bedded standard cage. Sleeping time was determined when the test animal was no longer capable to maintain its Righting Reflex. This reflex is defined as the animal' s failure of righting 2 times within 30 seconds. Clock time was recorded after the first administration of the test item and again when the animal woke up. The sleeping time was determined as the elapsed time between first administration of the tested item and the time that the animal regained its Righting Reflex. The entire study was recorded (VCR).
Observations for signs of morbidity and mortality were performed twice a day. One animal died immediately after diazepam dosing. Individual body weight determination was made on Day 1 prior to induction (see Table 16 and Fig. 5). At study termination all animals were euthanized by respiratory exposure to excess C02. Table 16. Body weights
Group Animal No. Body weight (g) Group Animal No. Body weight (g)
IF 1 183 2F 5 184
2 187 6 175
7 181
4 178 8 182
Average ± sd 182 ± 3.3 Average ± sd 180.5 ± 3.3
As shown in Table 17, sleeping time following drug treatment was clearly shortened. The large variation is due to the small number of animals in the two groups.
Table 17. Sleeping time
Group Animal No. Time (min) Group Animal No. Time (min)
IF 1 3.18 2F 5 40.46
2 5.24 6 10.1
- - 7 3.5
4 3.45 8 died
Average ± sd 3.96 ± 0.91 Average ± sd 18.02 ± 16.02
Example 9. Clinical study protocols
1. PK protocol
The primary objective is to determine the single dose absolute bioavailability of sublingual (SL) Formulation A using the marketed IV flumazenil formulation as the comparator. The secondary objectives are to characterize the concentration time course of two dose levels of SL Formulation A to support dose selection for Phase 2 and 3 studies and to evaluate the safety and tolerability of flumazenil formulations.
Study design includes masking and enrolment, open label, randomized, three-way crossover study, with 15 healthy subjects > 18 years of age, with no medication, based on the following study arms:
1. SL Formulation A 100 μΐ (1.1 mg) N=5
2. SL Formulation A 200 μΐ (2.2 mg) N=5
3. IV Flumazenil (Romazicon®) (0.2 mg) N=5
Study conditions: standardized as much as possible in terms of die and fluid intake. This type of study is interventional. The secondary outcome measure is to examine the safety and tolerability of Formulation A (l . lmg and 2.2mg), where the safety endpoints include: adverse event monitoring, vital signs, physical examinations, clinical laboratory tests and ECGs.
The time-points for blood and serum tests of flumazenil concentration are: 0, lOmin, 30min, 60min, 90min, 2h, 4h, 6h and 24h. PK parameters for analysis are Cmax, Tmax, Cmin, Tmin, AUC0-∞, AUC0-t and Ίυ2.
2. Phase 2B study: Dose range study of SL flumazenil
The study is a Phase 2B Dose Range, Efficacy and Safety of sublingual Formulation A for the indication: reversal of the residual effects of GABA related (benzodiazepine (BNZ) and nonBNZ) hypnotic drugs in insomniac patients. The residual sedative effect relates to benzodiazepine (BNZ) and nonBNZ hypnotic drugs. The study evaluates different doses of Formulation A in comparison to a placebo. Formulation A is provided as a sublingual (SL) spray formulation with flumazenil 1 lmg/ml.
Short-term safety and tolerability data is monitored along with psychomotor /cognitive and behavioral effects. The rationale of this study is to support study the pivotal efficacy phase 3 study, for the aforementioned indication.
Doses are determined with reference to the bioavailability study results. The study includes two groups of insomniac patients (males and females) aged >18y, Group A (n=60) and Group B (n=60). Each group is divided into Dose 1 (n=20), Dose 2 (n=20) and Placebo (n=20). The subjects all groups (n=20) are tested for the safety and efficacy of SL spray administration of Formulation A in the reversal of the sedative residual effect of BNZ (Group A) or Non BNZ (Group B).
Response to treatment is evaluated for each treatment arm. Good Response (GR) is R*25%, namely, an improvement of 25% in efficacy parameters of Formulation A treated patients compared to placebo/baseline. Very Good Response to treatment (VGR) is R*35%.
The secondary objectives are to evaluate the duration of action of a single dose of SL CRLS003 and to evaluate the safety of a single daily dose of Formulation A for 7 days. Study Design is randomized, double blind, dose range and placebo controlled. The following examinations are carried out:
a) Physical examination, clinical laboratory, vital signs.
b) Sleep induction by BNZ/nonBNZ (Study A/B, respectively), per patient routine.
c) All patients undergo full polysomnography.
d) At 6 AM patients are awaked.
e) Baseline - On awakening digit symbol substitution test (DSST) is performed.
f) Patients are randomly treated with Formulation A 1.1 or 2.2 mg or with placebo. g) The following tests are conducted at 10 min, 60 min, and 120 min following SL Formulation A or placebo administration: DSST, immediate Word Recall Test (iWRT), Visual Analog Scale (VAS) and Profile of Mood States (POMS).
h) Patients will be self-treated throughout the week at home using daily hypnotics and Formulation A/placebo (blinded) on awakening. Patients will answer questions and write sleep and performance logs.
i) On the 7th night, patients will report to the clinic for full polysomnography and will repeat the same evaluation as in the previous visit.
j) One week after termination, subjects will visit for safety monitoring.
Subjects meet all of the following inclusion criteria:
i. Each subject understands and voluntarily signs an informed consent form prior to any study-mandated procedure.
ii. Male or female aged >18y at screening. Women of childbearing potential must have a negative pregnancy test at the screening visit and use a reliable method of contraception during the entire study duration (e.g., contraceptive pill; intrauterine device; contraceptive injection (prolonged-release gestagen); subdermal implantation; vaginal ring or transdermal patch).
iii. Body mass index > 18.5 and < 32 kg/m2.
iv. Chronic insomniac who are regularly treated by BNZ (Group A) or nonBNZ (Group B) for at least 2 months and report residual morning symptoms.
v. Each subject is in good health as determined by a medical history, physical examination and ECG.
vi. Negative regarding any use of illicit drug, alcohol (ethanol), stimulants.
Exclusion criteria are: 1. Using medications, except hypnotics and contraceptives, 1 month prior to screening.
2. A change in the hypnotics within the last 2 months.
3. Any sleep associated complains other than insomnia.
4. History of Epilepsy and or anti-epileptic drugs.
5. Excessive caffeine consumption (> 500 mg per day).
6. Pregnancy or breast feeding.
7. Night shift workers within 1 month prior to the screening visit.
8. Clinically relevant ECG abnormalities.
9. History of alcohol or drug abuse within 3 years prior to the screening visit.
10. Cognitive Behavioral Therapy (CBT) started within 1 month prior to screening.
11. Known hypersensitivity to drugs of the same class as the study treatment, or any excipients of the drug formulation.
12. Treatment with another investigational drug within 1 month prior to the screening.
13. History of severe head injury.
The study includes the following visits:
Visit 1- Screening visit: A signed and dated informed consent form is collected from each participating subject, prior to any screening procedures. Screening visit takes place up to 14 days prior to treatment visit. Subjects who are screened are assigned a 3 -digit Screening number. During the screening visit, the following procedures are performed: Review Inclusion / Exclusion criteria; Medical history inquiry and demographic data; Record prior medication; Neurological assessment; Conduct physical examination including vital signs, height and weight measurements; Pregnancy test; 12-lead electrocardiography.
Visits 2 and 3 - Treatment at sleep laboratory visits: prior to Formulation A administration subjects undergo full physical examination, including sublingual and oral examination; vital signs measurements; adverse events and concomitant medication inquiry; Blood baseline tests. After determining that a subject is eligible, subjects receives a Randomization number and progresses to the sleep induction stage. At this stage the subject receives a sleep / hypnotic drug as follows:
Study A: BNZ (same drug and dose that patients use regularly)
Study B: nonBNZ (same drug and dose that patients use regularly)
Subjects sleep under full polysomnography until 6 AM. On awakening DSST is performed and the study Drug is administered: (At time 0) 0.1/0.2 ml Formulation A or placebo (randomly and blindly). Thereafter, at 10 min., 60 min., and 120 min. post Formulation A administration, subjects are tested for psychomotor performance and behavioral tasks and vital signs. Safety measurement are conducted after performance tasks completion, as follows, full physical examination, including the sublingual and the oral cavity testing; neurological assessment; vital signs measurements; AE inquiry and blood tests.
The participants are released by a physician and are not allowed to drive until 10 hours after hypnotic administration. On the 7th day after study visit 2, visit 3 is conducted applying same procedures as in visit plan 2.
Across the study at home, patients continue using hypnotics and, on awakening taking Formulation A. Patients also keep a daily sleep-wake-performance log. The following prohibitions apply to the visits: caffeine consumption after 2:00 pm on the day before any study visit; consumption of alcohol or grapefruit (including as juice) from the day prior to each of the study visits, and for 3 consecutive days; and napping on the day before any study visit. During study any diet or non-pharmacological activity are allowed if started at least 1 month prior to the screening visit and stable until 24 hours after the last administration of study treatment.
Follow-up visit- Subjects pay a safety visit at day 7 after treatment termination and are examined, including blood tests, and report adverse effects (AEs).
Unscheduled visit - Subjects are monitored for AEs throughout the study. For any reported AEs defined as "Moderate or "Severe", the subject is instructed to visit the clinic for safety measures as required. Causal AEs is monitored until resolved.
Study Duration: subjects participate in this study for 1 week of therapy and 1 week of follow-up. Interim analysis is carried (after 30 patients). Standard statistical methods are used to analyze the results (e.g. mean and standard deviation based on student-t test and ANOVA), where p <0.05 defines significance.
3. Phase 3 pivotal efficacy study
This study is a double blind evaluation of SL Formulation A for reversal of next day residual sedative effect from hypnotic drugs used to treat insomnia. It is directed to evaluate the safety and effectiveness of SL Formulation A in insomniac patients in reversing the next day residual effect of hypnotic drugs. The study is designed to collect short-term safety and tolerability data along with psycho motor/cognitive and behavioral function. It is an interventional type of study. The indication, drugs and treatment are similar to the those tested in the Phase 2B study.
Sample includes 300 subjects (150 per treatment group) randomized to flumazenil or placebo 1 : 1 ratio, stratified by BNZ or nonBNZ. Treatment response is evaluated for each treatment arm. Good Response (GR) is R*25%. Secondary objective include safety of SL Formulation A compared to placebo administered daily for 6 weeks. The Study Design is randomized, double blind, placebo and controlled.
All subjects are evaluated for the safety and efficacy of SL Formulation A to reverse the sedative effect of BNZ or nonBNZ applying the same routine used in Phase 2B study with the following exceptions:
a) Patients are self-treated for 3 weeks using daily hypnotics and Formulation A/placebo (blinded) on awakening,
b) By the end of the 3rd week ± 3 nights and at the last night (6 weeks after trial start), patients visit the clinic for full polysomnography and repeat the procedures and testing described above.
c) Two weeks after the last treatment, the patient take safety monitoring and AE inquiry.
Subjects eligible for the study meet all of the aforementioned inclusion and exclusion criteria. The study includes the same type of visits listed above, with the addition of a fourth sleep laboratory visit. 4. Phase 3 double blind placebo controlled trial for the evaluation of Formulation A in the treatment of Episodic Hepatic Encephalopathy (HE) grade >2.
The purpose of this study is to evaluate mental score/quality of life improvement in patients with episodic FIE following the administration of sublingual flumazenil ( Formulation A). This is an interventional type of study. The indication is: relief of cognitive impairment symptoms in patients with episodic HE. The investigational drug is Formulation A sublingual spray comprising 1.12mg /ml Flumazenil. The time to recovery from episodic HE grade >2 is defined as a decrease of Conn score to Grade < 2, or a decrease in Asterixis. The secondary objectives of this study are as follows:
a) Duration and number of HE-related hospitalization.
b) Time to any increase from baseline in Conn score.
c) Time to any increase from baseline in Asterixis grade
d) Number connection Test (NCT or Reitan Test)
e) Continuous reaction times to sound (CRT)
f) Block design test (BDT)
g) Symbol digit modalities test (SDMT)
h) Mean change from baseline in fatigue domain score on the Chronic Liver Disease Questionnaire (CLDQ)
The study design is randomized, placebo controlled, double blinded administered sublingual flumazenil twice a day for 3 weeks. During study, patients with episodic HE grade >2 are screened for a respond to flumazenil therapy. Baseline assessment includes in addition to the routine testing described below: Clinical assessment (physical and blood tests), EEG, plasma assays for benzodiazepines and NCT.
Physical and blood tests include :
a. Ammonia measurements, weight, renal functions electrolytes each visit.
b. EEG
c. A questionnaire regarding diarrhea and abdominal discomfort
d. Cognitive impairment
e. Performance memory test each visit (e.g. NCT and Reitan Test)
f. Quality of life evaluation (HE related hospitalization). Conn score and asterixis increase are documented.
The study population includes male and female, cirrhotic patients aged >18y, with episodic HE grade >2 .
Inclusion criteria that subjects eligible for enrolment into the study meet are:
a) Episodic HE grade >2.
b) Responsiveness to flumazenil therapy.
c) Each subject understands and voluntarily signs an informed consent form prior to any study-mandated procedure.
d) Male or female aged >18 at screening.
e) Women of childbearing potential must have a negative pregnancy test at the screening visit and use a reliable method of contraception during the entire study duration (e.g. Contraceptive pill; Intra-uterine device; Contraceptive injection (prolonged-release gestagen); Subdermal implantation; Vaginal ring or Transdermal patch).
Exclusion criteria include chronic BNZ therapy and epilepsy.
Safety assessment is based on changes from screening/baseline for clinical AEs reported by the subject, or observed by the investigator assessed by physical exam, oral and sublingual assessment, neurological assessment and vital signs.
Standard statistical methods are used to analyze the results (e.g. mean and standard deviation based on student-t test and ANOVA), where p <0.05 defines significance.
The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying current knowledge, readily modify and/or adapt for various applications such specific embodiments without undue experimentation and without departing from the generic concept, and, therefore, such adaptations and modifications should and are intended to be comprehended within the meaning and range of equivalents of the disclosed embodiments. It is to be understood that the phraseology or terminology employed herein is for the purpose of description and not of limitation. The means, materials, and steps for carrying out various disclosed functions may take a variety of alternative forms without departing from the invention.

Claims

A pharmaceutically acceptable complex of ethyl 8-fluoro-5,6-dihydro-5-methyl-6- oxo- 4H- imidazo [1,5-a] [1,4] benzodiazepine- 3-carboxylate or a salt thereof and a complexation agent or a derivative thereof, with the proviso that the complexation agent is other than cyclodextrin or meglumine.
The pharmaceutically acceptable complex of claim 1, wherein the complexation agent is an aromatic complexation agent.
The pharmaceutically acceptable complex of claim 1, wherein the complexation agent is selected from the group consisting of: nicotinamide, nicotinic acid, nicotinate, benzoic acid, benzoate, hydroxybenzoic acid, hydroxybenzoate, salicylic acid, salicylate, gentisic acid, gentisate, ethanolamide, toluic acid (ortho, meta or para), toluate (ortho, meta or para), aminobenzoic acid, aminobenzoate, anthranilic acid, anthranilate, butylmonoglycolsulfate and resorcinol.
The pharmaceutically acceptable complex of claim 3, wherein the complexation agent is selected from the group consisting of: nicotinamide, nicotinic acid, sodium nicotinate, potassium nicotinate, benzoic acid, sodium benzoate, potassium benzoate, hydroxybenzoic acid, sodium hydroxybenzoate, potassium hydroxybenzoate, salicylic acid, sodium salicylate, potassium salicylate, gentisic acid, sodium gentisate, potassium gentisate, ethanolamide, sodium toluate, toluic acid, potassium toluate, aminobenzoic acid, sodium aminobenzoate, potassium aminobenzoate, anthranilic acid, sodium anthranilate, potassium anthranilate, sodium butylmonoglycolsulfate, potassium butylmonoglycolsulfate and resorcinol.
The pharmaceutically acceptable complex of claim 3, wherein the complexation agent is selected from the group consisting of: nicotinamide, sodium nicotinate, sodium benzoate, sodium hydroxybenzoates, sodium salicylate, sodium gentisate, gentisic acid ethanolamide, sodium toluates, sodium aminobenzoate, sodium anthranilate, sodium butylmonoglycolsulfate and resorcinol.
The pharmaceutically acceptable complex of claim 3, wherein the complexation agent is nicotinamide or a derivative thereof.
The pharmaceutically acceptable complex of claim 4, wherein the complexation agent :flumazenil ratio is in the range of 1 : 1 to 2: 1.
8. The pharmaceutically acceptable complex of claim 4, which is a flumazenil nicotinamide complex.
9. A pharmaceutical composition comprising the flumazenil complex of any one of claims 1 to 8 and a pharmaceutical acceptable carrier.
10. The pharmaceutical composition according to claim 9, wherein the composition is in a form selected from the group consisting of: a pill, tablet, lozenge, coated tablet, granule, capsule, hard or soft gelatin capsule, aqueous solution, alcoholic solution, oily solution, syrup, emulsion suspension pastille suppository, solution for injection or infusion, ointment, tincture, cream, lotion, powder, spray, transdermal therapeutic systems, nasal spray, aerosol mixture, microcapsule, implant, rod and plaster.
11. The pharmaceutical composition according to claim 9, wherein the composition is in a form selected from the group consisting of: immediate release, delayed release, pulsatile release, continuous release and repetitive release.
12. The pharmaceutical composition according to claim 9, wherein the concentration of flumazenil is within the range of about 0.4 to 2 % w/w.
13. The pharmaceutical composition according to claim 12, wherein the concentration of flumazenil is within the range of about 0.5 to 1.8 % w/w.
14. The pharmaceutical composition according to claim 9, further comprising a solubilizing agent selected from an alcohol, a glycol and a combination thereof.
15. The pharmaceutical composition according to claim 14, wherein the solubilizing agent comprises a combination of an alcohol and a glycol and wherein the alcohol: glycol ratio is at least 1.5: 1.
16. The pharmaceutical composition according to claim 15, wherein the alcohol:glycol ratio is in the range of 1.5: 1 to 5: 1.
17. The pharmaceutical composition according to claim 14, wherein the solubilizing agent comprises a combination of an alcohol and a glycol and wherein the concentration of the solubilizing agent is at least 40%.
18. The pharmaceutical composition according to claim 17, wherein the concentration of the solubilizing agent is in the range of 40% to 60%.
19. The pharmaceutical composition according to claim 14, further comprising a buffering agent.
20. The pharmaceutical composition according to claim 14, further comprising at least one agent selected from the group consisting of: a penetration enhancer, a surfactant and cyclodextrin.
21. The pharmaceutical composition according to claim 14, further comprising a preservative selected from the group consisting of: benzyl alcohol, propylparaben, methylparaben and combinations thereof.
22. The pharmaceutical composition according to of claim 21, wherein the penetration enhancer is menthol.
23. The pharmaceutical composition according to of claim 19, wherein the buffering agent is selected from the group consisting of: citric buffer, sodium chloride and combination thereof.
24. The pharmaceutical composition according to of claim 20, wherein the surfactant is benzalkonium chloride.
25. A method for treating a disease or a disorder comprising administering to a patient in need thereof a flumazenil complex according to any one of claims 1-8 or a pharmaceutical composition comprising according to any one of claims 9 to 24, wherein said disease or disorder is selected from the group consisting of: excessive sleepiness, alcohol intoxication and hepatic encephalopathy
26. The method of claim 25, wherein the excessive sleepiness is associated with hypersomnia, drowsiness, overdose of a sleep drug, alcohol intoxication, sedation and anesthesia induced by a sleep drug, sleep drug addiction, stimulant addiction, Alzheimer's disease, anxiety, schizophrenia, the rebound effect of a sleep drug, balance impairment induced by a sleep drug or any combination thereof.
27. The method of claim 26, wherein the excessive sleepiness associated with alcohol intoxication is treated by any one or more of reversing the effects of alcohol intoxication, reducing the effects of alcohol intoxication, alleviating the effects of alcohol intoxication or improving performance after alcohol consumption.
28. The method of claim 25, wherein treating alcohol intoxication reverses the effects of alcohol intoxication, reduces the effects of alcohol intoxication, alleviates the effects of alcohol intoxication or improves performance after alcohol consumption.
29. The method of claim 25, wherein the flumazenil complex or pharmaceutical composition comprising said flumazenil complex is administered by a route selected from the group consisting of: oral, buccal, sublingual, transdermal, transmucosal, intranasal, intravenous, intraperitoneal, imtramuscular, subcutaneous and intrathecal.
30. The method of claim 25, further comprising co-administering a wakefulness promoting agent.
31. The method of claim 30, wherein the wakefulness promoting agent is selected from the group consisting of modafinil, armodafinil, adrafinil, methylphenidate, nefazodone, sodium oxybate, phentermine, pemoline, adrenaline, methylxantines, theobromine, caffeine and any combination thereof.
32. The method of claims 26, wherein the sleep drug is selected from the group consisting of a benzodiazepine, a benzodiazepine modulator, a benzodiazepine analog, a non- benzodiazepine, a 5-HT2A receptor antagonist, a melatonin receptor agonist, an orexin receptor antagonist, a selective serotonin reuptake inhibitor, an antihistamine and a herbal product.
33. The method of claim 32, wherein the sleep drug is a sedative/hypnotic drug.
34. The method of claim 32, wherein the sleep drug is a benzodiazepine sedative/hypnotic drug.
35. The method of claim 34, wherein the benzodiazepine is selected from the group consisting of alprazolam, bromazepam, clonazepam, clotiazepam, cloxazolam, diazepam, estazolam, etizolam, fludiazepam, flunitrazepam, flurazepam, halazepam, haloxazolam, lorazepam, medazepam, midazolam, nimetazepam, nitrazepam, olanzapine, oxazepam, quazepam, temazepam and triazolam.
36. The method of claim 32, wherein the sleep-drug is a non-benzodiazepine sedative/hypnotic drug.
37. The method of claim 36, wherein the non-benzodiazepine is selected from the group consisting of adipiplon, agomelatine, almoxerant, brotizolam, diphenhydramine, divaplon, doxepin, eplivanserin, doxylamine succinate, eszopiclone, indiplon, ocinaplon, pagoclone, pazinaclone, pruvanserin, suproclone, suriclone, L-tryptophan, 5-hydroxy-L-tryptophan, melatonin, melatonin receptor agonists, muramyl dipeptide, ramelteon, uridine, volinanserin, zaleplon, Zolpidem, imidazopyridine hypnotic Zolpidem, APD125, ACP-103, PD 200-390, HY10275, GW649863 and EVT-201.
38. The method of claim 32, wherein the step of treating excessive sleepiness comprises reversing benzodiazepine-induced sedation and anesthesia following therapeutic or diagnostic procedures.
39. Use of the flumazenil complex according to any one of claims 1 to 8, or a pharmaceutical composition according to any of claims 9 to 24, for the treatment of a disease selected from the group consisting of: excessive sleepiness, alcohol intoxication and hepatic encephalopathy.
40. A method for treating a disease or disorder associated with binding of an agent to the GABA receptor, comprising administering to a patient in need thereof a pharmaceutical composition comprising an active ingredient selected from flumazenil complex according to claims 1 to 8, or a pharmaceutical composition according to any of claims 9 to 24.
41. The method of claim 40, comprising the step of reversing the effect caused by the binding of the agent to the GABA receptor.
42. The method of claim 40, wherein the agent is a benzodiazepine or a non- benzodiazepine the binding of which to the GABA receptor causes sedation.
PCT/IL2012/050060 2011-02-23 2012-02-23 Flumazenil complexes, compositions comprising same and uses thereof WO2012114342A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CA2827336A CA2827336C (en) 2011-02-23 2012-02-23 Flumazenil complexes, compositions comprising same and uses thereof
RU2013139247/04A RU2013139247A (en) 2011-02-23 2012-02-23 FLUMAZENIL COMPLEXES CONTAINING THEIR COMPOSITIONS AND THEIR APPLICATIONS
CN201280019999.3A CN103502224A (en) 2011-02-23 2012-02-23 Flumazenil complexes, compositions comprising same and uses thereof
ES12749603T ES2702848T3 (en) 2011-02-23 2012-02-23 Flumazenil complexes, compositions comprising them and uses thereof
EP12749603.2A EP2678320B1 (en) 2011-02-23 2012-02-23 Flumazenil complexes, compositions comprising same and uses thereof
AU2012221704A AU2012221704B2 (en) 2011-02-23 2012-02-23 Flumazenil complexes, compositions comprising same and uses thereof
JP2013554977A JP2014511382A (en) 2011-02-23 2012-02-23 Flumazenil complex, composition containing it, and use thereof
IL227742A IL227742A (en) 2011-02-23 2013-07-31 Flumazenil complexes, compositions comprising same and uses thereof
US13/974,760 US9585893B2 (en) 2011-02-23 2013-08-23 Flumazenil complexes, compositions comprising same and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161445559P 2011-02-23 2011-02-23
US61/445,559 2011-02-23

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/974,760 Continuation US9585893B2 (en) 2011-02-23 2013-08-23 Flumazenil complexes, compositions comprising same and uses thereof

Publications (1)

Publication Number Publication Date
WO2012114342A1 true WO2012114342A1 (en) 2012-08-30

Family

ID=46720188

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2012/050060 WO2012114342A1 (en) 2011-02-23 2012-02-23 Flumazenil complexes, compositions comprising same and uses thereof

Country Status (9)

Country Link
US (1) US9585893B2 (en)
EP (1) EP2678320B1 (en)
JP (1) JP2014511382A (en)
CN (1) CN103502224A (en)
AU (1) AU2012221704B2 (en)
CA (1) CA2827336C (en)
ES (1) ES2702848T3 (en)
RU (1) RU2013139247A (en)
WO (1) WO2012114342A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2958594A4 (en) * 2013-02-22 2017-03-01 Eastgate Pharmaceuticals Inc. Pharmaceutical composition for enhanced transmucosal administration of benzodiazepines
US9585893B2 (en) 2011-02-23 2017-03-07 Coeruleus Ltd. Flumazenil complexes, compositions comprising same and uses thereof
US10688103B2 (en) 2014-04-15 2020-06-23 Balance Therapeutics, Inc. Methods for treating hypersomnia
US11065224B2 (en) 2016-07-22 2021-07-20 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
WO2022147318A1 (en) * 2020-12-31 2022-07-07 Arena Pharmaceuticals, Inc. Methods of treatment
US11400065B2 (en) 2019-03-01 2022-08-02 Flamel Ireland Limited Gamma-hydroxybutyrate compositions having improved pharmacokinetics in the fed state
US11504347B1 (en) 2016-07-22 2022-11-22 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11583510B1 (en) 2022-02-07 2023-02-21 Flamel Ireland Limited Methods of administering gamma hydroxybutyrate formulations after a high-fat meal
US11602512B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11602513B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11779557B1 (en) 2022-02-07 2023-10-10 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11986451B1 (en) 2016-07-22 2024-05-21 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US12128021B1 (en) 2024-04-23 2024-10-29 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10555952B2 (en) 2015-06-19 2020-02-11 Melt Pharmaceuticals, Inc. Pharmaceutical compositions and methods for anesthesiological applications
US10179136B2 (en) 2015-06-19 2019-01-15 Imprimis Pharmaceuticals, Inc. Pharmaceutical compositions and methods for anesthesiological applications
US10166240B2 (en) 2015-06-19 2019-01-01 Imprimis Pharmaceuticals, Inc. Pharmaceutical compositions and methods for anesthesiological applications
US20200297734A1 (en) 2015-06-19 2020-09-24 Melt Pharmaceuticals, Inc. Pharmaceutical compositions and methods for anesthesiological applications
US10391102B2 (en) 2015-06-19 2019-08-27 Melt Pharmaceuticals, Inc. Pharmaceutical compositions and methods for anesthesiological applications
KR101964571B1 (en) 2015-06-19 2019-04-02 해로우 헬스 인코포레이티드 Pharmaceutical composition for anesthesia
CN105596313B (en) * 2016-02-17 2018-06-29 新乡医学院第一附属医院 A kind of capsule preparations for treatment insomnia with reversed pharmacological function
WO2018156953A1 (en) 2017-02-24 2018-08-30 Nalu Medical, Inc. Apparatus with sequentially implanted stimulators
CN112245435B (en) * 2020-09-28 2023-06-13 湖南农业大学 Composition for rescuing gelsemium intoxication and application thereof
WO2022109052A1 (en) * 2020-11-18 2022-05-27 Bexson Biomedical, Inc. Flumazenil formulations for subcutaneous injection and methods of treatment using gaba receptor modulators
EP4284506A1 (en) * 2021-02-01 2023-12-06 Clear Scientific, Inc. Sequestration compounds for treatment of substance use disorder and uses thereof
CN114366714B (en) * 2021-04-29 2022-10-11 山东京卫制药有限公司 Agomelatine suspension nasal spray and application thereof

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4316839A (en) 1979-10-04 1982-02-23 Hoffman-La Roche Inc. Imidazodiazepine derivatives
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5570698A (en) 1995-06-02 1996-11-05 Siemens Corporate Research, Inc. System for monitoring eyes for detecting sleep behavior
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5682144A (en) 1995-11-20 1997-10-28 Mannik; Kallis Hans Eye actuated sleep prevention devices and other eye controlled devices
US5689241A (en) 1995-04-24 1997-11-18 Clarke, Sr.; James Russell Sleep detection and driver alert apparatus
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US6087353A (en) 1998-05-15 2000-07-11 Forbes Medi-Tech Inc. Phytosterol compositions and use thereof in foods, beverages, pharmaceuticals, nutraceuticals and the like
US20040186075A1 (en) 1998-02-23 2004-09-23 Thorsteinn Loftsson High-energy cyclodextrin complexes
WO2004089313A2 (en) 2003-04-01 2004-10-21 Transform Pharmaceuticals, Inc. Novel olanzapine forms and related methods of treatment
US7056934B2 (en) 2003-07-25 2006-06-06 Pfizer Inc Nicotinamide derivatives useful as PDE4 inhibitors
US7153870B2 (en) 2003-07-25 2006-12-26 Pfizer Inc. Nicotinamide derivatives useful as PDE4 inhibitors
US20070265193A1 (en) * 2004-05-25 2007-11-15 Pfizer Inc Tetraazabenzo[e]azulene derivatives and analogs thereof
WO2008071665A1 (en) 2006-12-12 2008-06-19 Smithkline Beecham Corporation A nicotinamide derivative useful as p38 kinase inhibitor
US20090130216A1 (en) * 2007-05-07 2009-05-21 Hale Biopharma Ventures Multimodal particulate formulations
WO2009114740A2 (en) 2008-03-12 2009-09-17 Emory University USE OF GABAa RECEPTOR ANTAGONISTS FOR THE TREATMENT OF EXCESSIVE SLEEPINESS AND DISORDERS ASSOCIATED WITH EXCESSIVE SLEEPINESS
US20090270373A1 (en) * 2008-04-11 2009-10-29 Cypress Biosciences, Inc. Treatment of down syndrom with benzodiazepine receptor antagonists
US20100130479A1 (en) * 2005-05-16 2010-05-27 Cook James M Gabaergic Agents to Treat Memory Deficits
WO2011073985A1 (en) * 2009-12-14 2011-06-23 Coeruleus Ltd. Compositions and methods of counteracting residual sedative effects of sleep/ hypnotic drugs

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3943137A (en) * 1973-07-09 1976-03-09 University Of Kansas Endowment Association Acronycine derivatives
US5811547A (en) 1992-10-14 1998-09-22 Nippon Shinyaju Co., Ltd. Method for inducing crystalline state transition in medicinal substance
GB9503601D0 (en) 1995-02-23 1995-04-12 Merck Sharp & Dohme Method of treatment and method of manufacture of medicament
US20030082107A1 (en) 1997-10-01 2003-05-01 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating an infectious disease or cancer
US20030077227A1 (en) 1997-10-01 2003-04-24 Dugger Harry A. Buccal, polar and non-polar spray or capsule containing drugs for treating disorders of the central nervous system
AU759280C (en) 1998-02-23 2004-01-22 Cyclops, Ehf High-energy cyclodextrin complexes
AU4100699A (en) 1998-05-28 1999-12-13 Sepracor, Inc. Compositions and methods employing r(-) fluoxetine and other active ingredients
US6716452B1 (en) 2000-08-22 2004-04-06 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US7919119B2 (en) 1999-05-27 2011-04-05 Acusphere, Inc. Porous drug matrices and methods of manufacture thereof
DK1180020T4 (en) 1999-05-27 2009-10-05 Acusphere Inc Porous drug matrices and processes for their preparation
US20010053776A1 (en) 2000-05-24 2001-12-20 Crawforth James Michael Tricyclic pyridin-2-one analogue as a GABA receptor ligand
AU2001281416A1 (en) 2000-07-19 2002-01-30 Pitmy International N.V. Transportation of nucleic acid substances
WO2003034980A2 (en) 2000-11-14 2003-05-01 New River Pharmaceuticals Inc. A novel pharmaceutical compound containing abacavir sulfate and methods of making and using same
US20020099013A1 (en) 2000-11-14 2002-07-25 Thomas Piccariello Active agent delivery systems and methods for protecting and administering active agents
BR0206549A (en) 2001-01-17 2004-02-25 Hythiam Inc Use of flumazenil in the production of a drug for the treatment of alcohol dependence
AU2002231811B2 (en) 2001-02-15 2006-02-16 Hythiam, Inc. Use of flumazenil to produce a medicament for the treatment of cocaine dependency
US6573237B2 (en) * 2001-03-16 2003-06-03 Eli Lilly And Company Protein formulations
US20030044458A1 (en) 2001-08-06 2003-03-06 Curtis Wright Oral dosage form comprising a therapeutic agent and an adverse-effect agent
US7985422B2 (en) 2002-08-05 2011-07-26 Torrent Pharmaceuticals Limited Dosage form
CN1674889A (en) 2002-08-19 2005-09-28 华晶基因技术有限公司 Methods of predicting GABAa receptor efficacies for flavonoid compounds and uses therefor
DE10304403A1 (en) 2003-01-28 2004-08-05 Röhm GmbH & Co. KG Process for the preparation of an oral dosage form with immediate disintegration and drug release
US20050137141A1 (en) 2003-10-24 2005-06-23 John Hilfinger Prodrug composition
WO2005063248A1 (en) 2003-12-22 2005-07-14 Sepracor Inc. Modafinil combination therapy for improving sleep quality
US20050164987A1 (en) 2003-12-24 2005-07-28 Barberich Timothy J. Melatonin combination therapy for improving sleep quality
US7727576B2 (en) 2004-04-16 2010-06-01 Hewlett-Packard Development Company, L.P. System and a method for producing layered oral dosage forms
US7900577B2 (en) 2004-04-27 2011-03-08 Hewlett-Packard Development Company, L.P. System and a method for starch-based, slow-release oral dosage forms
US20060167068A1 (en) 2005-01-26 2006-07-27 Seth Feuerstein Method of treating self-injurious behavior with glutamate modulating agents
KR20070112164A (en) 2005-02-15 2007-11-22 엘란 파마 인터내셔널 리미티드 Aerosol and injectable formulations of nanoparticulate benzodiazepine
WO2007009691A2 (en) 2005-07-15 2007-01-25 Laboratorios Del Dr. Esteve, S.A. Combination of substituted pyrazolines and anti -addictive agent
US7893040B2 (en) 2005-07-22 2011-02-22 Oculis Ehf Cyclodextrin nanotechnology for ophthalmic drug delivery
WO2008024490A2 (en) 2006-08-24 2008-02-28 Theraquest Biosciences, Inc. Oral pharmaceutical formulations of abuse deterrent cannabinoids and method of use
JP2010523716A (en) 2007-04-11 2010-07-15 サイプレス バイオサイエンシズ,インク. Treatment of Down's syndrome with benzodiazepine receptor antagonists
EP2065038A1 (en) 2007-11-30 2009-06-03 Pharnext New therapeutic approaches for treating Charcot-Marie-Tooth disease
WO2009124755A1 (en) 2008-04-08 2009-10-15 European Molecular Biology Laboratory (Embl) Compounds with novel medical uses and method of identifying such compounds
WO2009146388A1 (en) 2008-05-28 2009-12-03 The Trustees Of Columbia University In The City Of New York Voxel-based methods for assessing subjects using positron emission tomography
CN101322688B (en) * 2008-07-30 2010-07-21 海南本创医药科技有限公司 Flumazenil oil-in-water emulsion for vein and preparation thereof
US8802058B2 (en) 2010-04-19 2014-08-12 Gelmed, Llc Pharmaceutical compositions and methods for administering the same
ES2702848T3 (en) 2011-02-23 2019-03-05 Coeruleus Ltd Flumazenil complexes, compositions comprising them and uses thereof
US20140128379A1 (en) 2011-04-01 2014-05-08 Cpex Pharmaceuticals, Inc. Nasal formulations of benzodiazepine

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4316839A (en) 1979-10-04 1982-02-23 Hoffman-La Roche Inc. Imidazodiazepine derivatives
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5689241A (en) 1995-04-24 1997-11-18 Clarke, Sr.; James Russell Sleep detection and driver alert apparatus
US5570698A (en) 1995-06-02 1996-11-05 Siemens Corporate Research, Inc. System for monitoring eyes for detecting sleep behavior
US5682144A (en) 1995-11-20 1997-10-28 Mannik; Kallis Hans Eye actuated sleep prevention devices and other eye controlled devices
US20040186075A1 (en) 1998-02-23 2004-09-23 Thorsteinn Loftsson High-energy cyclodextrin complexes
US6087353A (en) 1998-05-15 2000-07-11 Forbes Medi-Tech Inc. Phytosterol compositions and use thereof in foods, beverages, pharmaceuticals, nutraceuticals and the like
WO2004089313A2 (en) 2003-04-01 2004-10-21 Transform Pharmaceuticals, Inc. Novel olanzapine forms and related methods of treatment
US7056934B2 (en) 2003-07-25 2006-06-06 Pfizer Inc Nicotinamide derivatives useful as PDE4 inhibitors
US7153870B2 (en) 2003-07-25 2006-12-26 Pfizer Inc. Nicotinamide derivatives useful as PDE4 inhibitors
US20070265193A1 (en) * 2004-05-25 2007-11-15 Pfizer Inc Tetraazabenzo[e]azulene derivatives and analogs thereof
US20100130479A1 (en) * 2005-05-16 2010-05-27 Cook James M Gabaergic Agents to Treat Memory Deficits
WO2008071665A1 (en) 2006-12-12 2008-06-19 Smithkline Beecham Corporation A nicotinamide derivative useful as p38 kinase inhibitor
US20090130216A1 (en) * 2007-05-07 2009-05-21 Hale Biopharma Ventures Multimodal particulate formulations
WO2009114740A2 (en) 2008-03-12 2009-09-17 Emory University USE OF GABAa RECEPTOR ANTAGONISTS FOR THE TREATMENT OF EXCESSIVE SLEEPINESS AND DISORDERS ASSOCIATED WITH EXCESSIVE SLEEPINESS
US20090270373A1 (en) * 2008-04-11 2009-10-29 Cypress Biosciences, Inc. Treatment of down syndrom with benzodiazepine receptor antagonists
WO2011073985A1 (en) * 2009-12-14 2011-06-23 Coeruleus Ltd. Compositions and methods of counteracting residual sedative effects of sleep/ hypnotic drugs

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ALS-NIELSEN B ET AL.: "Benzodiazepine receptor antagonists for hepatic encephalopathy", COCHRANE DATABASE OF SYSTEMATIC REVIEWS, no. 2, 2004
BOND AJ, CNS DRUGS, vol. 9, no. 1, 1998, pages 41 - 57
FONTI ET AL., NUCL MED BIOL., vol. 26, no. 6, 1999, pages 681 - 686
HODDES ET AL., PSYCHOPHYSIOLOGY, vol. 9, 1972, pages 150
JOHNS MW, SLEEP, vol. 14, no. 6, 1991, pages 540 - 545
KURAMOCHIA ET AL., BIOORGANIC & MEDICINAL CHEMISTRY, vol. 13, no. 12, 2005, pages 4022 - 4036
PATAT ET AL., CLIN PHARMACOL THER., vol. 56, no. 4, 1994, pages 430 - 436
RIZWAN ET AL., RECENT PAT DRUG DELIV FORMUL., vol. 3, no. 2, 2009, pages 105 - 124

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9585893B2 (en) 2011-02-23 2017-03-07 Coeruleus Ltd. Flumazenil complexes, compositions comprising same and uses thereof
EP2958594A4 (en) * 2013-02-22 2017-03-01 Eastgate Pharmaceuticals Inc. Pharmaceutical composition for enhanced transmucosal administration of benzodiazepines
US10688103B2 (en) 2014-04-15 2020-06-23 Balance Therapeutics, Inc. Methods for treating hypersomnia
US11826335B2 (en) 2016-07-22 2023-11-28 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11896572B2 (en) 2016-07-22 2024-02-13 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US12115142B2 (en) 2016-07-22 2024-10-15 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11504347B1 (en) 2016-07-22 2022-11-22 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US12115145B2 (en) 2016-07-22 2024-10-15 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11602512B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11602513B1 (en) 2016-07-22 2023-03-14 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11766418B2 (en) 2016-07-22 2023-09-26 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US12115144B2 (en) 2016-07-22 2024-10-15 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11065224B2 (en) 2016-07-22 2021-07-20 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11839597B2 (en) 2016-07-22 2023-12-12 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US12115143B2 (en) 2016-07-22 2024-10-15 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11986451B1 (en) 2016-07-22 2024-05-21 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US12097176B2 (en) 2016-07-22 2024-09-24 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US12097175B2 (en) 2016-07-22 2024-09-24 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US12109186B2 (en) 2016-07-22 2024-10-08 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11400065B2 (en) 2019-03-01 2022-08-02 Flamel Ireland Limited Gamma-hydroxybutyrate compositions having improved pharmacokinetics in the fed state
WO2022147318A1 (en) * 2020-12-31 2022-07-07 Arena Pharmaceuticals, Inc. Methods of treatment
US11779557B1 (en) 2022-02-07 2023-10-10 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics
US11583510B1 (en) 2022-02-07 2023-02-21 Flamel Ireland Limited Methods of administering gamma hydroxybutyrate formulations after a high-fat meal
US12128021B1 (en) 2024-04-23 2024-10-29 Flamel Ireland Limited Modified release gamma-hydroxybutyrate formulations having improved pharmacokinetics

Also Published As

Publication number Publication date
EP2678320A4 (en) 2014-12-24
US9585893B2 (en) 2017-03-07
CA2827336A1 (en) 2012-08-30
ES2702848T3 (en) 2019-03-05
AU2012221704A2 (en) 2013-12-05
AU2012221704A1 (en) 2013-09-19
EP2678320A1 (en) 2014-01-01
AU2012221704B2 (en) 2013-12-05
CN103502224A (en) 2014-01-08
US20130345202A1 (en) 2013-12-26
CA2827336C (en) 2016-01-26
RU2013139247A (en) 2015-03-27
EP2678320B1 (en) 2018-09-19
JP2014511382A (en) 2014-05-15

Similar Documents

Publication Publication Date Title
US9585893B2 (en) Flumazenil complexes, compositions comprising same and uses thereof
US10912744B2 (en) Therapeutic formulation for reduced drug side effects
ES2738678T3 (en) Orvepitant for the treatment of chronic cough
US20170105983A1 (en) Compositions for reduction of side effects
US20120295893A1 (en) Compositions and methods of counteracting residual sedative effects of sleep/ hypnotic drugs
JP6258264B2 (en) Compositions and methods for increasing human platelet levels
US9066949B2 (en) Compositions and methods for the treatment of catatonia
IL227742A (en) Flumazenil complexes, compositions comprising same and uses thereof
JP2019536769A (en) Mucosal agent delivery
KR20240006600A (en) dosing regimen
BR112019018700A2 (en) pharmaceutical compositions and their uses

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12749603

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2013554977

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2827336

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2012221704

Country of ref document: AU

Date of ref document: 20120223

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2013139247

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2012749603

Country of ref document: EP