WO2009134877A2 - Therapeutics for treatment resistant mental disorders - Google Patents

Therapeutics for treatment resistant mental disorders Download PDF

Info

Publication number
WO2009134877A2
WO2009134877A2 PCT/US2009/042101 US2009042101W WO2009134877A2 WO 2009134877 A2 WO2009134877 A2 WO 2009134877A2 US 2009042101 W US2009042101 W US 2009042101W WO 2009134877 A2 WO2009134877 A2 WO 2009134877A2
Authority
WO
WIPO (PCT)
Prior art keywords
htt
mice
alkyl
alkenyl
derivative
Prior art date
Application number
PCT/US2009/042101
Other languages
French (fr)
Other versions
WO2009134877A3 (en
Inventor
Lynette Daws
Original Assignee
Board Of Regents, The University Of Texas System
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 Board Of Regents, The University Of Texas System filed Critical Board Of Regents, The University Of Texas System
Publication of WO2009134877A2 publication Critical patent/WO2009134877A2/en
Publication of WO2009134877A3 publication Critical patent/WO2009134877A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/133Amines having hydroxy groups, e.g. sphingosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse

Definitions

  • the invention generally relates to therapeutic agents for the treatment of mood disorders.
  • the invention generally relates to the use of organic cation transporter blockers for the treatment of mood disorders, including but not limited to depression, as well as related orders such as alcoholism.
  • Serotonin (5-HT) neurotransmission is tightly regulated by high affinity uptake of released 5-HT by the 5-HT transporter (5-HTT or SERT, herein referred to as 5-HTT).
  • 5-HTT is also a major site of action for many psychotherapeutic and addictive drugs.
  • a common deletional polymorphism in the promoter region (5-HTTLPR) of the human 5-HTT gene confers low 5-HTT expression. Carriers of this short (s) gene variant are more prone to psychiatric disorders and are often resistant to conventional treatment with selective 5-HT reuptake inhibitors (SSRIs) compared to individuals homozygous for the long (1) allele.
  • SSRIs selective 5-HT reuptake inhibitors
  • OCTs Organic cation transporters
  • a role for OCTs in regulating central serotonergic neurotransmission has remained unexplored.
  • OCTs are widely expressed in brain, that they are corticosterone-, methamphetamine- and methylenedioxymethamphetamine (MDMA)- sensitive and that their blockade robustly increases extracellular 5-HT in brain.
  • MDMA methylenedioxymethamphetamine
  • a method of inhibiting or reducing the effects of a mental disorder in a subject includes administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising an alkylamino-catechol derivative.
  • a method of inhibiting or reducing the effects of a mental disorder in a subject includes administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising a quinoline derivative.
  • a method of inhibiting or reducing the effects of a mental disorder in a subject includes administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising a bis-quinoline derivative.
  • FIG. IA depicts histograms of the amount of 0CT3 protein expressed in mice genetically modified to exhibit no (5-HTT-/-) or half as many 5-HTTs (5-HTT+ ⁇ ) as a wildtype mice (5- HTT+/+). From herein 5-HTT+/- and 5-HTT-/- mice are referred to as 5-HTT mutant mice or genetically modified mice;
  • FIG. IB depicts histograms of the relationship between 5-HTT and 0CT3 expression in the hippocampus of genetically modified mice;
  • FIGS. 2A and 2B depict plots of the effect of decynium-22 on the inhibition of 5-HT clearance
  • FIG. 2C depicts a plot of the effect of corticosterone on the inhibition of 5-HT clearance
  • FIG. 3A depicts a histogram of the clearance time of histamine from genetically modified mice
  • FIG. 3B depicts a histogram of the clearance time of histamine in the presence of decynium-22 from genetically modified mice;
  • FIG. 4 depicts a histogram of the time spent immobile of genetically modified mice in a tail suspension test in the presence and absence of decynium-22. While the invention may be susceptible to various modifications and alternative forms, specific embodiments thereof are shown by way of example in the drawings and will herein be described in detail. The drawings may not be to scale. It should be understood, however, that the drawings and detailed description thereto are not intended to limit the invention to the particular form disclosed, but to the contrary, the intention is to cover all modifications, equivalents, and alternatives falling within the spirit and scope of the present invention as defined by the appended claims.
  • OCTs are functionally up-regulated in 5-HTT heterozygote and KO mice. For example, 0CT3 expression and OCT-mediated clearance of 5- HT are increased in a manner inversely proportional to 5-HTT expression. Furthermore, OCT blockade has potent antidepressant-like activity in 5-HTT mutant mice. OCTs are therefore a novel target for the development of new therapeutics for the treatment of affective disorders, particularly in individuals resistant to treatment with SSRIs.
  • the hippocampus is an important brain structure mediating the therapeutic response to treatment with antidepressants and is a region where effects of antidepressant drugs have been extensively studied.
  • OCT 1 and 3 are located in neurons and glia in the hippocampus of mice and rats. 0CT3 expression is greater than that of OCTl in the hippocampus.
  • the relationship between OCT and 5-HTT expression may be studied by quantifying levels of mRNA using quantitative RTPCR. 5-HTT gene expression was determined to be dependent on 5-HTT genotype.
  • Table 1 OCTl and OCT3 mRNA expression in hippocampus
  • Figure IB illustrates the reciprocal relationship between 5-HTT and OCT3 expression in hippocampus of 5-HTT mutant mice, providing further evidence that OCT3 function may be increased in response to reduced 5-HTT expression.
  • OCT3 The distribution of OCT3 in the hippocampus was determined to confirm its expression in regions of the hippocampus where SSRIs have potent actions to inhibit 5-HT clearance and to aid in the selection of stereotaxic coordinates for placement of carbon fiber electrodes used for high-speed chronoamperometric recordings of 5-HT clearance in vivo. It was found that OCT3 was located most densely in the pyramidal and granule cell layers of the hippocampus and appeared to be contained primarily within the neuronal cell bodies. Additionally, differences in the distribution of OCT3 immunostaining in the hippocampus were not detected among 5-HTT genotypes indicating that the cellular distribution of OCT3 does not change as a function of 5- HTT genotype. In wildtype and 5-HTT+/- mice, 5-HTTs are also expressed in these hippocampal layers. Thus, OCT3 is also positioned in the proximity of 5-HT terminals and could increase its function when 5-HTT expression and/or function is compromised.
  • D-22 Prior to use in vivo, the effect of the OCT3 blocker D-22, on the electrochemical properties of the Nafion-coated carbon fiber electrodes used for chronoamperometric recordings was tested. D-22 itself did not produce an oxidation current, however, when applied in amounts exceeding 1.0 micromolar the sensitivity of the carbon fiber to detect 5-HT was reduced, likely due to adherence of D-22 to the surface of the electrode. Because of this, dose-response analysis was limited to 1.4 pmol (pressure-ejection of 140 nl of 10 micromolar D-22).
  • the concentration of drug reaching the recording electrode is estimated to be between 10- and 200-fold more dilute than the concentration in the micropipette, thus by not exceeding a barrel concentration of 10 micromolar, the sensitivity of the carbon fiber electrode for 5-HT was preserved.
  • 5-HTT+/- and 5-HTT-/- mice the 5-HT signal returned to pre-drug values within 30 minutes following application of D-22.
  • Signal amplitude was not significantly different among genotypes before drug (0.51 + 0.09 ⁇ M, 0.53 + 0.05 ⁇ M and 0.54 + 0.05 ⁇ M for 5-HTT+/+, 5-HTT+/- and 5-HTT-/- mice respectively).
  • D-22 did not significantly affect signal amplitude in any genotype, although there was a trend for the amplitude to be greater in 5-HTT-/- mice two minutes after D-22 (0.60 + 0.04 ⁇ M) compared to baseline signal amplitude.
  • OCTs are capable of taking up 5-HT from extracellular fluid but is revealed only when 5-HTT expression is compromised.
  • D-22 has an appreciable affinity for the plasma membrane monoamine transporter (PMAT), which can also transport 5-HT.
  • PMAT plasma membrane monoamine transporter
  • 5-HT clearance was examined in the hippocampus in response to locally-applied corticosterone (55 pmol), which blocks OCT3 but not PMAT.
  • 5-HT is the major substrate for the 5-HTT, it is also a substrate for OCT and other transporters including NET and DAT. Histamine is a substrate for OCT3 but not other biogenic amine transporters. Therefore, to isolate OCT-mediated uptake from that mediated by 5-HTT and other transporters, we measured clearance of histamine from the CA3 region of hippocampus.
  • D-22 1.0 ⁇ g/kg
  • the dose of D-22, 1.0 ⁇ g/kg, used in our study was chosen based on pilot studies to identify a dose that did not produce directly observable effects that may interfere with TST performance. At a higher dose of 1.0 mg/kg, D-22 decreased locomotion. To our knowledge, there is only one published report documenting behavioral effects of D-22. In this study D-22 was administered directly into the medial hypothalamus of rats via a microdialysis probe. These authors reported an increase in the amount of time the rats spent grooming, but no other overt behavioral effects were observed.
  • OCT3 becomes an important player in regulation of 5- HT neurotransmission when 5-HTT expression is low or absent.
  • implications for OCTs in mediating fundamental physiologic processes are vastly more far-reaching than regulating extracellular 5-HT alone.
  • OCTs orchestrate "normal" physiological and neurological function.
  • OCTs play an important role in central nervous system regulation of salt intake, and the stress axis as well as their regulation by chronic treatment with the psychostimulant methamphetamine.
  • a polymorphism of the gene encoding OCT3 was recently linked to methamphetamine use disorder.
  • OCT3 As a potential target for new antidepressant treatments as well as the potential to be used in the treatment of alcoholism.
  • OCT3 appears to be an integral player in regulating 5-HT neurotransmission in the face of reduced 5-HTT expression or function.
  • OCT3 may be an effective target for drug development in the treatment of psychiatric disorders that is better tailored to the individual.
  • OCTs are polyspecific, transporting a host of cationic molecules including neurotransmitters (e.g. norepinephrine, dopamine, histamine). They are also inhibited by hormones including corticosterone, progesterone and testosterone.
  • OCTs Drugs found to interact with OCTs that may be used as an alternate antidepressant treatment include, but are not limited to, receptor agonists and antagonists, ion channel and transporter blockers, anesthetics, antibiotics, antiviral agents, antidiabetic agents and psychostimulants.
  • OCTs are expressed in most organs, with particularly high expression in kidney, liver, heart, lung and brain. Thus, they are positioned to play a significant role in the development and/or treatment of numerous diseases including hypertension, cardiac dysfunction and mental disorders. For individuals whose genetic make-up imposes reduced 5-HTT expression, 0CT3 expression and function increases as a compensatory alternative for 5-HT uptake.
  • Alcoholism is one of the burdensome of neuropsychiatric disorders in modern society, yet remains poorly understood and ineffectively treated. Alcohol acts at many and varied target proteins in brain to influence neurotransmission.
  • the serotonin (5-HT) system has been strongly linked with susceptibility to alcoholism.
  • Reduced 5-HTT expression elevates risk for alcoholism.
  • expression and function of the 0CT3, which also transports 5-HT is upregulated in the brain of mice genetically manipulated to constitutively reduce 5-HTT expression.
  • mice with reduced 5-HTT expression were more sensitive to the sedative effects of alcohol as well as its ability to inhibit clearance of 5-HT from extracellular fluid.
  • amino-catechol derivatives may be used to treat mental disorders such as depression.
  • amino-catechol derivatives may be used to treat mental disorders, particularly for patients that exhibit limited response to 5-HTT inhibiting drugs, lamino-catechol derivatives have the general structure (I):
  • R 1 is hydrogen, alkyl, or -COOH
  • R 2 and R 3 are hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl
  • each R 4 is independently hydrogen, alkyl, alkenyl, aryl or benzyl.
  • an amino-catechol derivative has the structure (II):
  • R 1 is hydrogen, alkyl, or -COOH; where R 2 is alkyl, cycloalkyl, alkenyl, or akynyl; and where R 4 is independently hydrogen, alkyl, alkenyl, aryl or benzyl.
  • alkylamino-catechol derivatives that may be used for the treatment of mental disorders include, but are not limited to, normetanephrine, 4-hydroxy-3- methoxyphenylserine, and 3-O-methylisoprenaline.
  • a quinoline derivative may be used to treat mental disorders such as depression.
  • quinoline derivatives may be used to treat mental disorders, particularly for patients that exhibit limited response to 5-HTT inhibiting drugs.
  • Quinoline derivatives have the general structure (HI):
  • each R 5 is independently hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl and
  • R 6 is hydrogen, alkyl, alkenyl, akynyl, or -CO 2 R 5
  • cyanine 863 An example of a quinoline derivative that may be used for the treatment of mental disorders is cyanine 863.
  • bis-quinoline derivatives may be used to treat mental disorders such as depression.
  • bis-quinoline derivatives may be used to treat mental disorders, particularly for patients that exhibit limited response to 5-HTT inhibiting drugs.
  • Bis- quinoline derivatives have the general structures (IV) and (V):
  • each R is independently hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl
  • An example of a bis-quinoline derivative of structure (III) that may be used for the treatment of mental disorders is decynium-22 ("DEC-22", both R 5 are ethyl).
  • An example of a bis-quinoline derivative of structure (IV) that may be used for the treatment of mental disorders is decynium- 24 ("DEC-24", both R 5 are ethyl).
  • bisquinolines include compounds (IVa) and (Va)
  • any pharmaceutically acceptable salts of the compounds having structures (III) and (IV) may be used. It should be understood that treatment of mental disorders may be achieved by use of an effective amount of one or more alkylamine-catechol derivatives, quinoline derivatives, bis- quinoline derivatives, or combinations thereof.
  • alkyl generally refers to a chemical substituent containing carbon and hydrogen only (a hydrocarbon) without any double or triple bonds being present.
  • Alkyl includes branched or unbranched monovalent saturated hydrocarbon radicals. Examples of alkyl groups include, but are not limited to: methyl; ethyl; n-propyl; isopropyl; n-butyl; iso- butyl; sec-butyl; tert-butyl; n-pentyl; 1-methylbutyl; 2-methylbutyl; 3-methylbutyl; 2,2- dimethylpropyl; 1-ethylpropyl; 1,1-dimethylpropyl; and 1,2-dimethylpropyl.
  • cycloalkyl generally refers to a chemical substituent containing carbon and hydrogen only (a hydrocarbon) without any double or triple bonds being present where the hydrocarbon is in a form that includes one or more rings.
  • the rings may be unsubstituted or substituted with alkyl groups.
  • cycloalkyl groups include, but are not limited to: cyclopropyl; cyclopropylmethyl; cyclopropylethyl; cyclobutyl; cyclobutylmethyl; cyclobutylethyl; cyclopentyl; cyclopentylmethyl; cyclopentylethyl, and cyclohexyl.
  • Alkenyl groups include alkenyl groups substituted with: alkyl substituents or cycloalkyl substituents; or alkyl groups or cycloalkyl groups substituted with an alkenyl group.
  • alkenyl groups include, but are not limited to: vinyl; allyl; 2-cyclopropyl-l-ethenyl; 1-propenyl; 1-butenyl; 2-butenyl; 3-butenyl; 2-methyl-l-propenyl; 2- methyl-2-propenyl; 1-pentenyl; 2-pentenyl; 3-pentenyl; 4-pentenyl; 3 -methyl- 1-butenyl; 3- methyl-2-butenyl; 3-methyl-3-butenyl; 2-methyl- 1-butenyl; 2-methyl-2-butenyl; 2-methyl-3- butenyl; 2-ethyl-2-propenyl; 1 -methyl- 1-butenyl; l-methyl-2-butenyl; and l-methyl-3-butenyl.
  • alkynyl generally refers to any structure or moiety having the unsaturation C ⁇ C.
  • Alkynyl groups include alkynyl groups substituted with: alkyl substituents or cycloalkyl substituents; or alkyl groups or cycloalkyl groups substituted with an alkynyl group.
  • alkynyl groups include, but are not limited to: ethynyl; 1-propynyl; 2-propynyl; 1-butynyl; 2- butynyl; 3-butynyl; 1-pentynyl; 2-pentynyl; 3-pentynyl; 4-pentynyl; l-methyl-2-propynyl; 1- methyl-2-butynyl; l-methyl-3-butynyl; 2-methyl-3-butynyl; l,2-dimethyl-3-butynyl; and 2,2- dimethyl- 3- butynyl.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • Pharmaceutically acceptable acid addition salts of the compounds of the invention include salts derived from inorganic acids such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorus, and the like, as well as the salts derived from organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc.
  • inorganic acids such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorus, and the like
  • organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc.
  • Such salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, phthalate, benzenesulfonate, toluenesulfonate, phenylacetate, citrate, lactate, maleate, tartrate, methanesulfonate, and the like.
  • salts of amino acids such as arginate, gluconate, galacturonate, and the like; see, for example, Berge et al., "Pharmaceutical Salts," J. of Pharmaceutical Science, 1977; 66:1 19.
  • the acid addition salts of the basic compounds are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt in the conventional manner.
  • the free base form may be regenerated by contacting the salt form with a base, and isolating the free base in the conventional manner.
  • the free base forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free base for purposes of the present invention.
  • Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metal hydroxides, or of organic amines.
  • metals used as cations are sodium, potassium, magnesium, calcium, and the like.
  • suitable amines are N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine, and procaine; see, for example, Berge et al., supra., 1977.
  • the base addition salts of acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner.
  • the free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in a conventional manner.
  • the free acid forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acid for purposes of the present invention. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
  • compositions described herein may be used to treat a subject.
  • Any suitable route of administration may be employed for providing a patient with an effective dosage of compounds described above or pharmaceutically acceptable salts thereof.
  • oral, rectal, topical, parenteral, ocular, intracranial, pulmonary, nasal, and the like may be employed.
  • Dosage forms may include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, oils, emulsions, liposomes, aerosols, and the like.
  • the compositions described herein be administered parenterally.
  • compositions may include those compositions suitable for oral, rectal, topical, parenteral (including subcutaneous, intramuscular, and intravenous), ocular (ophthalmic), pulmonary (aerosol inhalation), or nasal administration, although the most suitable route in any given case will depend on the nature and severity of the conditions being treated and on the nature of the active ingredient. They may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy. For administration by inhalation, the compositions described herein are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or nebulizers. The compositions may also be delivered as powders which may be formulated and the powder composition may be inhaled with the aid of an insufflation powder inhaler device. Suitable topical formulations for use in the present embodiments may include transdermal devices, aerosols, creams, ointments, lotions, dusting powders, and the like.
  • compositions can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous).
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, capsules and tablets, with the solid oral preparations being preferred over the liquid preparations. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques.
  • the pharmaceutical preparations may be manufactured in a manner which is itself known to one skilled in the art, for example, by means of conventional mixing, granulating, dragee- making, softgel encapsulation, dissolving, extracting, or lyophilizing processes.
  • pharmaceutical preparations for oral use may be obtained by combining the active compounds with solid and semi- solid excipients and suitable preservatives, and/or co-antioxidants.
  • the resulting mixture may be ground and processed.
  • the resulting mixture of granules may be used, after adding suitable auxiliaries, if desired or necessary, to obtain tablets, softgels, lozenges, capsules, or dragee cores.
  • Suitable excipients may be fillers such as saccharides (e.g., lactose, sucrose, or mannose), sugar alcohols (e.g., mannitol or sorbitol), cellulose preparations and/or calcium phosphates (e.g., tricalcium phosphate or calcium hydrogen phosphate).
  • binders may be used such as starch paste (e.g., maize or corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone).
  • Disintegrating agents may be added (e.g., the above-mentioned starches) as well as carboxymethyl- starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof (e.g., sodium alginate).
  • Auxiliaries are, above all, flow-regulating agents and lubricants (e.g., silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol, or PEG).
  • Dragee cores are provided with suitable coatings, which, if desired, are resistant to gastric juices.
  • Softgelatin capsules are provided with suitable coatings, which, typically, contain gelatin and/or suitable edible dye(s).
  • animal component-free and kosher gelatin capsules may be particularly suitable for the embodiments described herein for wide availability of usage and consumption.
  • concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol (PEG) and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures, including dimethylsulfoxide (DMSO), tetrahydrofuran (THF), acetone, ethanol, or other suitable solvents and co-solvents.
  • DMSO dimethylsulfoxide
  • THF tetrahydrofuran
  • acetone acetone
  • ethanol or other suitable solvents and co-solvents.
  • cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethyl-cellulose phthalate
  • Dye stuffs or pigments may be added to the tablets or dragee coatings or softgelatin capsules, for example, for identification or in order to characterize combinations of active compound doses, or to disguise the capsule contents for usage in clinical or other studies.
  • Other pharmaceutical preparations that may be used orally include push-fit capsules made of gelatin, as well as soft, thermally sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol.
  • the push-fit capsules may contain the active compounds in the form of granules that may be mixed with fillers such as, for example, lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers and/or preservatives.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils such as rice bran oil or peanut oil or palm oil, or liquid paraffin.
  • suitable liquids such as fatty oils such as rice bran oil or peanut oil or palm oil, or liquid paraffin.
  • stabilizers and preservatives may be added.
  • pulmonary administration of a pharmaceutical composition may be desirable.
  • Pulmonary administration may include, for example, inhalation of aerosolized or nebulized liquid or solid particles of the pharmaceutically active component dispersed in and surrounded by a gas.
  • Possible pharmaceutical preparations which may be used rectally or vaginally, include, for example, suppositories, which include a combination of the active compounds with a suppository base.
  • Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons.
  • gelatin rectal capsules that consist of a combination of the active compounds with a base.
  • Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
  • Suitable formulations for parenteral administration include, but are not limited to, aqueous solutions of the active compounds in water-soluble and/or water dispersible form, for example, water-soluble salts, esters, carbonates, phosphate esters or ethers, sulfates, glycoside ethers, together with spacers and/or linkers.
  • Suspensions of the active compounds as appropriate oily injection suspensions may be administered, particularly suitable for intramuscular injection.
  • Suitable lipophilic solvents, co-solvents (such as DMSO or ethanol), and/or vehicles including fatty oils, for example, rice bran oil or peanut oil and/or palm oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides, may be used.
  • Aqueous injection suspensions may contain substances that increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol, dextran, and/or cyclodextrins. Cyclodextrins (e.g., ⁇ - cyclodextrin) may be used specifically to increase the water solubility for parenteral injection of the compound.
  • Liposomal formulations in admixture with, for example, egg yolk phosphotidylcholine (E-PC), may be made for injection.
  • the suspension may contain stabilizers, for example, antioxidants such as BHT, and/or preservatives, such as benzyl alcohol.
  • compositions of this invention can be administered in such oral dosage forms as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. They may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
  • the dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.
  • a physician or veterinarian may determine and prescribe the effective amount of the drug required to prevent, counter, or arrest the progress or the development prostate cancer in a subject.
  • the pharmaceutical compositions may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four or more times daily.
  • compositions described herein may further be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using transdermal skin patches.
  • suitable intranasal vehicles or via transdermal routes, using transdermal skin patches.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as "pharmacologically inert carriers") suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices.
  • the pharmacologically active component may be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like;
  • an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like
  • the oral drug components can be combined with any oral, nontoxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
  • suitable binders, lubricants, disintegrating agents, and coloring agents can also be incorporated into the mixture.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • the compounds of the present invention may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine, or phosphatidylcholines.
  • Compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers.
  • soluble polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues.
  • the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
  • a drug for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
  • Gelatin capsules may contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
  • powdered carriers such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
  • Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • water, a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions.
  • Solutions for parenteral administration preferably contain a water-soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances.
  • Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents.
  • citric acid and its salts and sodium EDTA are also used.
  • parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propyl-paraben, and chlorobutanol.
  • the pharmaceutical composition may be administered to the patient systemically.
  • systemic includes subcutaneous injection; intravenous, intramuscular, intraestemal injection; infusion; inhalation, transdermal administration, oral administration; and intra-operative instillation.
  • respirable particles comprising the active compound, which the subject inhales.
  • the active compound would be absorbed into the bloodstream via the lungs, and subsequently contact the lacrimal glands in a pharmaceutically effective amount.
  • the respirable particles may be liquid or solid, with a particle size sufficiently small to pass through the mouth and larynx upon inhalation; in general, particles ranging from about 1 to 10 microns, but more preferably 1-5 microns, in size are considered respirable.
  • Another method of systemically administering the active compounds involves administering a liquid/liquid suspension in the form of eye drops or eye wash or nasal drops of a liquid formulation, or a nasal spray of respirable particles that the subject inhales.
  • Liquid pharmaceutical compositions of the active compound for producing a nasal spray or nasal or eye drops may be prepared by combining the active compound with a suitable vehicle, such as sterile pyrogen free water or sterile saline by techniques known to those skilled in the art.
  • the active compounds may also be systemically administered through absorption by the skin using transdermal patches or pads.
  • the active compounds are absorbed into the bloodstream through the skin.
  • Plasma concentration of the active compounds can be controlled by using patches containing different concentrations of active compounds.
  • systemic administration of the active compound involves oral administration, in which pharmaceutical compositions containing active compounds are in the form of tablets, lozenges, aqueous or oily suspensions, viscous gels, chewable gums, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Additional means of systemic administration of the active compound to the subject may involve a suppository form of the active compound, such that a therapeutically effective amount of the compound reaches the eyes via systemic absorption and circulation. Further means of systemic administration of the active compound involve direct intraoperative instillation of a gel, cream, or liquid suspension form of a therapeutically effective amount of the active compound.
  • the solution containing the active compound may contain a physiologically compatible vehicle, as those skilled in the art can select, using conventional criteria.
  • the vehicles may be selected from the known pharmaceutical vehicles which include, but are not limited to, saline solution, water polyethers such as polyethylene glycol, polyvinyls such as polyvinyl alcohol and povidone, cellulose derivatives such as methylcellulose and hydroxypropyl methylcellulose, petroleum derivatives such as mineral oil and white petrolatum, animal fats such as lanolin, polymers of acrylic acid such as carboxypolymethylene gel, vegetable fats such as peanut oil and polysaccharides such as dextrans, and glycosaminoglycans such as sodium hyaluronate and salts such as sodium chloride and potassium chloride.
  • water polyethers such as polyethylene glycol
  • polyvinyls such as polyvinyl alcohol and povidone
  • cellulose derivatives such as methylcellulose and hydroxypropyl methylcellulose
  • petroleum derivatives such as mineral oil and white petrol
  • the pharmaceutical formulation is prepared in a sterile medium.
  • the active ingredient depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle.
  • Adjuvants such as local anaesthetics, preservatives and buffering agents can also be dissolved in the vehicle.
  • the sterile injectable preparation may be a sterile injectable solution or suspension in a non-toxic acceptable diluent or solvent.
  • acceptable vehicles and solvents that may be employed are sterile water, saline solution, or Ringer's solution.
  • the OCT3 inhibitor used may be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous).
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, capsules and tablets, with the solid oral preparations being preferred over the liquid preparations. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques.
  • 5-HTT mutant mice were generated according to the method of described in Bengel D, et al. (1998) "Altered brain serotonin (5-HT) homeostasis and locomotor insensitivity to MDMA ("ecstasy") in 5-HT transporter deficient mice.” MoI Pharmacol 53:649-655. Male, 5-HTT+/+, 5 -HTT+/-, or 5-HTT -/- mice, weighing 25 to 30 g, were used for all experiments. RT-PCR was used to quantify OCT 1 and 3 mRNA according to the method of Chen J-J, et al.
  • Tissue was assayed for protein content by the method of Bradford M-M (1976) "A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein- dye-binding.
  • Immunocytochemical staining for OCT3 was performed using the same antibody as used for Western blot analyses.
  • High-speed chronoamperometry was used according to established protocols (Daws L-C, Toney G-M (2007) "Voltammetric Methods to Study Kinetics and Mechanisms for Serotonin Clearance In Vivo.”
  • Electrochemical Methods in Neuroscience for Methods and New Frontiers in Neuroscience, eds Michael A-C, Simon S-A, Nicolelis M-A-L.
  • mice were injected intraperitoneally with either D-22 (1.0 ⁇ g/kg) or saline and then placed in an observation chamber for 30 min. Immediately following this period they were securely fastened by the distal end of the tail to a flat metallic surface and suspended in a visually isolated area (40 x 40 x 40 cm white box). The amount of time spent immobile, defined as the absence of limb movement, was recorded over a 6-min test session by a trained observer who remained blind to genotype and treatment. The mice underwent a second TST 60 min. following injection of D-22 or saline. The effects of genotype and drugs were analyzed using ANOVA followed by Bonferroni post-hoc tests. Data are presented as the mean and standard error of the mean (s.e.m.).

Landscapes

  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Neurology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Psychiatry (AREA)
  • Addiction (AREA)
  • Pain & Pain Management (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Described herein are treatments for patients suffering from a mental disorders that include administering an effective amount of an alkylamine-catechol derivative, a quinoline derivative, a bis-quinoline derivative, or combinations thereof.

Description

THERAPEUTICS FOR TREATMENT RESISTANT MENTAL DISORDERS
REFERENCE TO GOVERNMENT SPONSORED RESEARCH
This invention was made with U.S. government support under grant no. MH064489 awarded by the National Institutes of Health. The U.S. government has certain rights in the invention.
BACKGROUND
1. Field of the Invention The invention generally relates to therapeutic agents for the treatment of mood disorders.
More particularly, the invention generally relates to the use of organic cation transporter blockers for the treatment of mood disorders, including but not limited to depression, as well as related orders such as alcoholism.
2. Description of the Relevant Art Dysfunction of the serotonergic system is strongly linked to many psychiatric illnesses, ranging from affective disorders to drug abuse and alcoholism. Serotonin (5-HT) neurotransmission is tightly regulated by high affinity uptake of released 5-HT by the 5-HT transporter (5-HTT or SERT, herein referred to as 5-HTT). The 5-HTT is also a major site of action for many psychotherapeutic and addictive drugs. A common deletional polymorphism in the promoter region (5-HTTLPR) of the human 5-HTT gene confers low 5-HTT expression. Carriers of this short (s) gene variant are more prone to psychiatric disorders and are often resistant to conventional treatment with selective 5-HT reuptake inhibitors (SSRIs) compared to individuals homozygous for the long (1) allele.
It is believed that the upregulation of an alternate 5-HT uptake mechanism might explain reduced clinical efficacy of SSRIs in carriers of the s allele. Related to this, we have discovered that ethanol increases extracellular 5-HT in the CA3 region of the hippocampus by inhibiting 5- HT clearance. Surprisingly, however, ethanol inhibition of 5-HT clearance was most pronounced in mice lacking the 5-HTT (5-HTT-/- mice) or with reduced 5-HTT expression (5- HTT+/- mice). These results unmasked the existence of a novel mechanism for 5-HT clearance and most significantly, are consistent with the idea that novel transporters for 5-HT may undergo adaptive up-regulation when 5-HTT expression or function is compromised. If so, then they also provide a neurochemical basis for resistance to treatment with SSRIs. Specifically, the buffering imposed by a functionally up-regulated alternate transporter for 5-HT may prevent extracellular 5-HT from climbing to levels sufficiently high to trigger the adaptive neurochemical events necessary for therapeutic benefit.
The identity of such a transporter is yet to be determined. Some efforts have focused on the closely related norepinephrine and dopamine transporters (NET and DAT), which can both transport 5-HT. However, while there is evidence that DAT is a compensatory alternative for 5- HT uptake in certain brain regions of mice that lack the 5-HTT there is no evidence supporting such a role for the NET, at least in the C A3 region of hippocampus; the region where recordings of OCT function described herein, were made.
Organic cation transporters (OCTs) also have a high capacity for 5-HT uptake, albeit with a lower affinity than the 5-HTT. However, until very recently a role for OCTs in regulating central serotonergic neurotransmission has remained unexplored. We now know that OCTs are widely expressed in brain, that they are corticosterone-, methamphetamine- and methylenedioxymethamphetamine (MDMA)- sensitive and that their blockade robustly increases extracellular 5-HT in brain. Of particular note is evidence that expression of at least some OCT subtypes is increased in 5-HTT KO mice.
SUMMARY OF THE INVENTION
In an embodiment, a method of inhibiting or reducing the effects of a mental disorder in a subject includes administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising an alkylamino-catechol derivative. In another embodiment, a method of inhibiting or reducing the effects of a mental disorder in a subject includes administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising a quinoline derivative.
In another embodiment, a method of inhibiting or reducing the effects of a mental disorder in a subject includes administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising a bis-quinoline derivative.
BRIEF DESCRIPTION OF THE DRAWINGS
Advantages of the present invention will become apparent to those skilled in the art with the benefit of the following detailed description of embodiments and upon reference to the accompanying drawings in which: FIG. IA depicts histograms of the amount of 0CT3 protein expressed in mice genetically modified to exhibit no (5-HTT-/-) or half as many 5-HTTs (5-HTT+Λ) as a wildtype mice (5- HTT+/+). From herein 5-HTT+/- and 5-HTT-/- mice are referred to as 5-HTT mutant mice or genetically modified mice; FIG. IB depicts histograms of the relationship between 5-HTT and 0CT3 expression in the hippocampus of genetically modified mice;
FIGS. 2A and 2B depict plots of the effect of decynium-22 on the inhibition of 5-HT clearance; FIG. 2C depicts a plot of the effect of corticosterone on the inhibition of 5-HT clearance; FIG. 3A depicts a histogram of the clearance time of histamine from genetically modified mice; FIG. 3B depicts a histogram of the clearance time of histamine in the presence of decynium-22 from genetically modified mice; and
FIG. 4 depicts a histogram of the time spent immobile of genetically modified mice in a tail suspension test in the presence and absence of decynium-22. While the invention may be susceptible to various modifications and alternative forms, specific embodiments thereof are shown by way of example in the drawings and will herein be described in detail. The drawings may not be to scale. It should be understood, however, that the drawings and detailed description thereto are not intended to limit the invention to the particular form disclosed, but to the contrary, the intention is to cover all modifications, equivalents, and alternatives falling within the spirit and scope of the present invention as defined by the appended claims.
DETAILED DESCRIPTION
Our studies have determined that OCTs are functionally up-regulated in 5-HTT heterozygote and KO mice. For example, 0CT3 expression and OCT-mediated clearance of 5- HT are increased in a manner inversely proportional to 5-HTT expression. Furthermore, OCT blockade has potent antidepressant-like activity in 5-HTT mutant mice. OCTs are therefore a novel target for the development of new therapeutics for the treatment of affective disorders, particularly in individuals resistant to treatment with SSRIs.
The hippocampus is an important brain structure mediating the therapeutic response to treatment with antidepressants and is a region where effects of antidepressant drugs have been extensively studied. Of three OCT subtypes (OCT 1, 2 and 3), OCT 1 and 3 are located in neurons and glia in the hippocampus of mice and rats. 0CT3 expression is greater than that of OCTl in the hippocampus. The relationship between OCT and 5-HTT expression may be studied by quantifying levels of mRNA using quantitative RTPCR. 5-HTT gene expression was determined to be dependent on 5-HTT genotype. In hippocampus of 5-HTT+/- mice, 5-HTT mRNA levels were about half (43 + 10% n=5; t6 = 2.882, P = 0.028) that of 5-HTT+/+ mice and were undetectable in 5-HTT knockout mice (data not shown). OCTl gene expression in the hippocampus did not differ among 5-HTT genotypes, although there was a trend for higher levels of mRNA in 5 -HTT-/- compared to 5-HTT+/+ 5 mice (Table 1). However, as others have reported, we found that mRNA for OCT3 was increased (+28%) in hippocampus of 5-HTT-/- mice compared to 5-HTT+/+ mice (t9 = 2.686, P = 0.025). We extended these findings to show that OCT3 mRNA levels were also significantly elevated (+34%) in 5HTT+/- mice compared to wildtype mice (til = 2.85, P = 0.0158) (Table 1).
Figure imgf000005_0001
Table 1: OCTl and OCT3 mRNA expression in hippocampus
values expressed as mean + s.e.m. mRNA/ 18SmRNA - relative units
*P = 0.025; **P = 0.0158, number of mice per group shown in parentheses
Western blot analysis was carried out to determine if increased levels of mRNA in 5- HTT mutant mice were accompanied by corresponding increases in protein expression. Figure IA shows that OCT3 protein is increased in 5-HTT+/- (+27%) and 5-HTT-/- (+36%) mice relative to 5-HTT+/+ mice (5-HTT+/- P = 0.0276; 5-HTT-/- P = 0.0063, versus 5-HTT+/+, 1- way ANOVA with Tukey's post-hoc test, n=9/genotype). Hippocampal OCTl protein expression did not vary among 5-HTT genotypes (OCTl/β-actin expression ratios were 0.834 + 0.037, 0.847 + 0.045 and 0.799 + 0.059 for 5-HTT+/+, 5-HTT+/- and 5-HTT-/- mice respectively n=6/genotype). These data indicate that OCT3 is a candidate for regulation of 5-HT clearance in mice with reduced or no 5-HTTs. Figure IB illustrates the reciprocal relationship between 5-HTT and OCT3 expression in hippocampus of 5-HTT mutant mice, providing further evidence that OCT3 function may be increased in response to reduced 5-HTT expression.
The distribution of OCT3 in the hippocampus was determined to confirm its expression in regions of the hippocampus where SSRIs have potent actions to inhibit 5-HT clearance and to aid in the selection of stereotaxic coordinates for placement of carbon fiber electrodes used for high-speed chronoamperometric recordings of 5-HT clearance in vivo. It was found that OCT3 was located most densely in the pyramidal and granule cell layers of the hippocampus and appeared to be contained primarily within the neuronal cell bodies. Additionally, differences in the distribution of OCT3 immunostaining in the hippocampus were not detected among 5-HTT genotypes indicating that the cellular distribution of OCT3 does not change as a function of 5- HTT genotype. In wildtype and 5-HTT+/- mice, 5-HTTs are also expressed in these hippocampal layers. Thus, OCT3 is also positioned in the proximity of 5-HT terminals and could increase its function when 5-HTT expression and/or function is compromised.
Having established that OCT3 expression is increased in the hippocampus of 5-HTT+/- and 5-HTT-/- mice, we determined that the potency of OCT3 blockers (e.g. decynium-22, "D- 22"), to inhibit 5-HT clearance would be greater in these mutants than in 5-HTT+/+ mice. Highspeed chronoamperometry can be used to measure clearance of 5-HT locally applied into the CA3 region of hippocampus of anesthetized mice, before and after intrahippocampal application of D-22. The CA3 region was selected based on our finding that OCT3 is expressed in this region and because the norepinephrine and dopamine transporters do not contribute significantly to 5-HT clearance under conditions used here. Prior to use in vivo, the effect of the OCT3 blocker D-22, on the electrochemical properties of the Nafion-coated carbon fiber electrodes used for chronoamperometric recordings was tested. D-22 itself did not produce an oxidation current, however, when applied in amounts exceeding 1.0 micromolar the sensitivity of the carbon fiber to detect 5-HT was reduced, likely due to adherence of D-22 to the surface of the electrode. Because of this, dose-response analysis was limited to 1.4 pmol (pressure-ejection of 140 nl of 10 micromolar D-22). The concentration of drug reaching the recording electrode is estimated to be between 10- and 200-fold more dilute than the concentration in the micropipette, thus by not exceeding a barrel concentration of 10 micromolar, the sensitivity of the carbon fiber electrode for 5-HT was preserved. Highlighted in Figure 2A & 2B is the 5-HTT genotype- dependency of the effect of D-22 to inhibit 5-HT clearance. Serotonin was pressure-ejected into the CA3 region to generate reproducible signals and then D-22 or an equivalent volume of vehicle (phosphate buffered saline (PBS)) was applied directly into the same region via a glass multi-barrel pipette. Representative traces are shown in Figure 2A. D-22 slowed 5-HT clearance in a concentration-dependent (F3,58 = 11.93; P = 0.0116) and genotype dependent
(F2,58 = 11.00; P = 0.0062) manner. As evident in Figure 2B there was also a shift to the left in the dose-effect relationship in 5-HTT-/- mice, relative to 5-HTT+/- and 5-HTT+/+ mice. Clearance of 5-HT in 5-HTT-/- mice was significantly inhibited by D-22 at both 0.14 and 1.4 pmol amounts (P < 0.001), whereas only the highest dose (1.4 pmol) of D-22 produced significant inhibition in 5-HTT+/- mice (P < 0.05). In 5-HTT+/+ mice, D-22 did not affect 5-HT clearance at any of the pmol amounts tested. Vehicle was without effect on 5-HT clearance in all genotypes. Consistent with our published findings, baseline clearance times (T80, the time for the 5-HT signal to decline by eighty percent of the peak amplitude) were longer in 5-HTT-/- mice (110 + 5 s, n = 5), compared to 5-HTT+/- (83 + 14 s, n = 6) and 5-HTT+/+ mice (75 + 8 s, n = 8) and were not different between 5-HTT+/+ and 5-HTT+/- mice. Two minutes after intrahippocampal D-22 (1.4 pmol), Tgo values increased further in 5-HTT-/- (156 + 29 s) and 5- HT+/- (97 + 13 s) mice, but not in 5-HTT+/+ (74 + 5 s) mice. In 5-HTT+/- and 5-HTT-/- mice, the 5-HT signal returned to pre-drug values within 30 minutes following application of D-22. Signal amplitude was not significantly different among genotypes before drug (0.51 + 0.09 μM, 0.53 + 0.05 μM and 0.54 + 0.05 μM for 5-HTT+/+, 5-HTT+/- and 5-HTT-/- mice respectively). D-22 did not significantly affect signal amplitude in any genotype, although there was a trend for the amplitude to be greater in 5-HTT-/- mice two minutes after D-22 (0.60 + 0.04 μM) compared to baseline signal amplitude.
Together, the present data indicate that OCTs are capable of taking up 5-HT from extracellular fluid but is revealed only when 5-HTT expression is compromised. However, in addition to its action at the OCT, D-22 has an appreciable affinity for the plasma membrane monoamine transporter (PMAT), which can also transport 5-HT. To distinguish OCT-mediated 5-HT uptake from that mediated by PMAT, 5-HT clearance was examined in the hippocampus in response to locally-applied corticosterone (55 pmol), which blocks OCT3 but not PMAT. As shown in Figure 2C, there was again a significant effect of genotype, with no effect of corticosterone in 5-HTT+/+ mice, and the magnitude of inhibition increasing in a fashion inversely proportional to the number of 5-HTTs, being greatest in 5-HTT-/- mice (F2,15 = 8.609; P = 0.0032). These data suggest that the effect of D-22 and corticosterone to inhibit 5-HT clearance is mediated by OCT and not PMAT.
Although 5-HT is the major substrate for the 5-HTT, it is also a substrate for OCT and other transporters including NET and DAT. Histamine is a substrate for OCT3 but not other biogenic amine transporters. Therefore, to isolate OCT-mediated uptake from that mediated by 5-HTT and other transporters, we measured clearance of histamine from the CA3 region of hippocampus. Consistent with our evidence that OCT3 expression and function is increased in 5-HTT mutants, histamine clearance was faster in 5-HTT+/- (76 + 13 s, n = 6) and 5 -HTT-/- mice (68 + 11 s, n = 8) than in 5-HTT+/+ mice (142 + 22 s, n = 6) (F2.17 = 6.814; P = 0.0067; Figure 3.A.). Likewise, the effect of D-22 to inhibit histamine clearance was significantly greater in 5-HTT mutant mice than wildtype mice (F2.17 = 4.146; P = 0.0342; Figure 3.B). One way that antidepressant drugs are thought to initiate therapeutic efficacy is by increasing extracellular 5-HT. The robust inhibitory effect of the OCT3 blocker, D-22, on 5-HT clearance in the CA3 region of hippocampus in 5-HTT mutant mice, indicates that an OCT3 blocker (such as D-22) may have antidepressant efficacy in these mice. We, therefore, measured the time spent immobile in the tail suspension test (TST), a commonly used paradigm to test drugs for antidepressant activity. We reasoned that because 5-HT clearance was slowed after use of an OCT3 blocker (e.g., D-22) in 5-HTT+/- and 5-HTT-/- mice (and therefore extracellular 5- HT levels elevated from basal, regardless of what that level might be), we might expect an antidepressant- like effect of OCT3 blockers in 5-HTT mutant mice. In contrast, because OCT3 blockers did not change 5-HT clearance in 5-HTT+/+ mice, we hypothesized that OCT3 blockers would have little, if any, antidepressant- like effect in this 5-HTT genotype. Time spent immobile was significantly decreased 30 min. after D-22 (1.0 μg/kg, intraperitoneally) compared to saline treated mice (Fl, 42 = 10.05, P = 0.0034). However, the decrease in immobility time only reached statistical significance in 5-HTT-/- mice. In a second TST, 60 min. after injection, immobility time remained less in D-22-treated 5-HTT mutant mice compared to saline control mice (F1.42 = 12.76, P = 0.0121). The antidepressant-like effect of D-22 persisted in 5-HTT-/- mice (P < 0.05) and became significant in 5-HTT+/- mice. Again, D-22 treatment did not result in any significant change in immobility time in 5-HTT+/+ mice. Figure 4 illustrates results for the TST carried out 60 min. post injection of D-22 or saline where data within each genotype are presented as a percent of immobility time of saline-treated mice. Antidepressant-like effects of D-22 were apparent in 5-HTT mutant mice but absent in wildtype mice (main effect treatment: Fl, 42 = 15.01, P = 0.0052). It is important to note that the baseline phenotypes of these mice in the TST are consistent with those of researchers, showing no difference between 5-HTT genotypes (bred on a C57B1/6 background as we are using) in the amount of time spent immobile. All genotypes spent approximately half of the 6-minute test period immobile. For example, time spent immobile in the test 60 min. following injection of saline was 167 + 13 s, 164 + 17 s and 179 + 16 s for 5-HTT+/+, +/- and -/- mice respectively (n = 8 per group). By comparison, after D-22 the time spent immobile was 164 + 16 s, 125 + 27 s and 125 + 16 s in 5- HTT+/+, +/- and -/- mice respectively (n=8 per group).
It has been found that the tricyclic antidepressant and NET blocker desipramine (DMI) decreased the time spent immobile in the TST to a greater extent in 5-HTT-/- mice than in 5- HTT+/+ and 5-HTT+/- mice. One explanation may be that NET upregulates in 5-HTT-/- mice, but interestingly DMI has appreciable affinity for OCTs (IC50 values ranging from 5-14 μM). Taken together with the present data, OCTs might well contribute to this greater antidepressant- like effect of DMI in 5-HTT-/- mice. Along these lines it is noteworthy that mice subjected to acute footshock 2-4 min. prior to the TST spend less time immobile than control mice. The footshock resulted in a dramatic increase in plasma corticosterone. Thus, the antidepressant- like effect of acute stress might also be explained by acute blockade of 0CT3 by corticosterone. Supporting this idea, a more recent study reported that elevation of plasma corticosterone, by including the hormone in drinking water of mice, also produced an antidepressant- like effect in the forced swim test.
The dose of D-22, 1.0 μg/kg, used in our study was chosen based on pilot studies to identify a dose that did not produce directly observable effects that may interfere with TST performance. At a higher dose of 1.0 mg/kg, D-22 decreased locomotion. To our knowledge, there is only one published report documenting behavioral effects of D-22. In this study D-22 was administered directly into the medial hypothalamus of rats via a microdialysis probe. These authors reported an increase in the amount of time the rats spent grooming, but no other overt behavioral effects were observed. Over a range of doses tested in pilot studies we selected 1.0 μg/kg D-22 for our studies using the TST as this dose induced a modest amount of grooming compared to saline-injected mice and thereby demonstrated that D-22 was indeed acting centrally. Importantly, locomotor activity measured for 30 min. after injection of D-22 or saline was not different among genotypes, nor did it differ as a function of treatment. It is unlikely therefore, that increased general motor activity accounts for the decreased immobility time of 5- HTT+/- and 5-HTT-/- mice in the TST after D-22 treatment.
Our data clearly demonstrate that OCT3 becomes an important player in regulation of 5- HT neurotransmission when 5-HTT expression is low or absent. However, it should be emphasized that the implications for OCTs in mediating fundamental physiologic processes are vastly more far-reaching than regulating extracellular 5-HT alone. We believe that OCTs orchestrate "normal" physiological and neurological function. For example, OCTs play an important role in central nervous system regulation of salt intake, and the stress axis as well as their regulation by chronic treatment with the psychostimulant methamphetamine. Related to this, a polymorphism of the gene encoding OCT3 was recently linked to methamphetamine use disorder. Our data, together with others, point to OCT3 as a potential target for new antidepressant treatments as well as the potential to be used in the treatment of alcoholism. Specifically, OCT3 appears to be an integral player in regulating 5-HT neurotransmission in the face of reduced 5-HTT expression or function. Thus it has been found that OCT3 may be an effective target for drug development in the treatment of psychiatric disorders that is better tailored to the individual. From a more general perspective, OCTs are polyspecific, transporting a host of cationic molecules including neurotransmitters (e.g. norepinephrine, dopamine, histamine). They are also inhibited by hormones including corticosterone, progesterone and testosterone. Drugs found to interact with OCTs that may be used as an alternate antidepressant treatment include, but are not limited to, receptor agonists and antagonists, ion channel and transporter blockers, anesthetics, antibiotics, antiviral agents, antidiabetic agents and psychostimulants. OCTs are expressed in most organs, with particularly high expression in kidney, liver, heart, lung and brain. Thus, they are positioned to play a significant role in the development and/or treatment of numerous diseases including hypertension, cardiac dysfunction and mental disorders. For individuals whose genetic make-up imposes reduced 5-HTT expression, 0CT3 expression and function increases as a compensatory alternative for 5-HT uptake. Individuals with low 5-HTT expression, such as carriers of the s allele, are also more prone to psychiatric disorders and more resistant to treatment with SSRIs than are individuals homozygous for the long (1) allele. This may be, at least in part, attributed to increased 0CT3 expression and function. Because SSRIs are currently the first line treatment for depression and related disorders, individuals who respond poorly to SSRIs have presented a major challenge to health care providers and significant efforts have been made to develop improved therapeutics. Data presented herein, provide evidence that OCTs, 0CT3 in particular, are rational targets for the development of new therapeutics to treat depression and related disorders, such as alcoholism, in SSRI resistant patients.
Alcoholism is one of the burdensome of neuropsychiatric disorders in modern society, yet remains poorly understood and ineffectively treated. Alcohol acts at many and varied target proteins in brain to influence neurotransmission. The serotonin (5-HT) system has been strongly linked with susceptibility to alcoholism. Reduced 5-HTT expression elevates risk for alcoholism. As noted herein expression and function of the 0CT3, which also transports 5-HT, is upregulated in the brain of mice genetically manipulated to constitutively reduce 5-HTT expression. Moreover, it has been found that mice with reduced 5-HTT expression were more sensitive to the sedative effects of alcohol as well as its ability to inhibit clearance of 5-HT from extracellular fluid. In one test, co-administration of an OCT blocker, together with alcohol, produced no net additive effect to inhibit 5-HT clearance. This result suggests that alcohol does indeed act on OCTs and that blockers of this transporter (i.e., OCT inhibitors) are useful treatments for alcoholism in their own right, or as add-ons to current medications used to treat alcoholism. In one embodiment, amino-catechol derivatives may be used to treat mental disorders such as depression. For example, amino-catechol derivatives may be used to treat mental disorders, particularly for patients that exhibit limited response to 5-HTT inhibiting drugs, lamino-catechol derivatives have the general structure (I):
Figure imgf000011_0001
where R1 is hydrogen, alkyl, or -COOH; where R2 and R3 are hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl; and where each R4 is independently hydrogen, alkyl, alkenyl, aryl or benzyl.
In another embodiment, an amino-catechol derivative has the structure (II):
Figure imgf000011_0002
(H)
where R1 is hydrogen, alkyl, or -COOH; where R2 is alkyl, cycloalkyl, alkenyl, or akynyl; and where R4 is independently hydrogen, alkyl, alkenyl, aryl or benzyl.
Examples of alkylamino-catechol derivatives that may be used for the treatment of mental disorders include, but are not limited to, normetanephrine, 4-hydroxy-3- methoxyphenylserine, and 3-O-methylisoprenaline.
Figure imgf000011_0003
normetanephrine 4-hydroxy-3-methoxyphenylserine
Figure imgf000012_0001
3-O-methylisoprenaline
It should also be understood that any pharmaceutically acceptable salts of the compounds having structure (I) and (II) may be used.
In another embodiment, a quinoline derivative may be used to treat mental disorders such as depression. For example, quinoline derivatives may be used to treat mental disorders, particularly for patients that exhibit limited response to 5-HTT inhibiting drugs. Quinoline derivatives have the general structure (HI):
Figure imgf000012_0002
(πi)
where each R5 is independently hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl and
R6 is hydrogen, alkyl, alkenyl, akynyl, or -CO2R5
An example of a quinoline derivative that may be used for the treatment of mental disorders is cyanine 863.
Figure imgf000013_0001
cyanine 863
It should also be understood that any pharmaceutically acceptable salts of the compounds having structure (II) may be used.
In another embodiment, bis-quinoline derivatives may be used to treat mental disorders such as depression. For example, bis-quinoline derivatives may be used to treat mental disorders, particularly for patients that exhibit limited response to 5-HTT inhibiting drugs. Bis- quinoline derivatives have the general structures (IV) and (V):
Figure imgf000013_0002
(IV) (V)
where each R is independently hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl
An example of a bis-quinoline derivative of structure (III) that may be used for the treatment of mental disorders is decynium-22 ("DEC-22", both R5 are ethyl). An example of a bis-quinoline derivative of structure (IV) that may be used for the treatment of mental disorders is decynium- 24 ("DEC-24", both R5 are ethyl).
Figure imgf000013_0003
decynium-22 decynium-24
Other bisquinolines include compounds (IVa) and (Va)
Figure imgf000014_0001
(IVa) (Va)
It should also be understood that any pharmaceutically acceptable salts of the compounds having structures (III) and (IV) may be used. It should be understood that treatment of mental disorders may be achieved by use of an effective amount of one or more alkylamine-catechol derivatives, quinoline derivatives, bis- quinoline derivatives, or combinations thereof.
The term "alkyl" as used herein generally refers to a chemical substituent containing carbon and hydrogen only (a hydrocarbon) without any double or triple bonds being present. Alkyl includes branched or unbranched monovalent saturated hydrocarbon radicals. Examples of alkyl groups include, but are not limited to: methyl; ethyl; n-propyl; isopropyl; n-butyl; iso- butyl; sec-butyl; tert-butyl; n-pentyl; 1-methylbutyl; 2-methylbutyl; 3-methylbutyl; 2,2- dimethylpropyl; 1-ethylpropyl; 1,1-dimethylpropyl; and 1,2-dimethylpropyl.
The term "cycloalkyl" as used herein generally refers to a chemical substituent containing carbon and hydrogen only (a hydrocarbon) without any double or triple bonds being present where the hydrocarbon is in a form that includes one or more rings. The rings may be unsubstituted or substituted with alkyl groups. Examples of cycloalkyl groups include, but are not limited to: cyclopropyl; cyclopropylmethyl; cyclopropylethyl; cyclobutyl; cyclobutylmethyl; cyclobutylethyl; cyclopentyl; cyclopentylmethyl; cyclopentylethyl, and cyclohexyl. The terms "alkenyl" and "alkene" as used herein generally refer to any structure or moiety having the unsaturation C=C. Alkenyl groups include alkenyl groups substituted with: alkyl substituents or cycloalkyl substituents; or alkyl groups or cycloalkyl groups substituted with an alkenyl group. Examples of alkenyl groups include, but are not limited to: vinyl; allyl; 2-cyclopropyl-l-ethenyl; 1-propenyl; 1-butenyl; 2-butenyl; 3-butenyl; 2-methyl-l-propenyl; 2- methyl-2-propenyl; 1-pentenyl; 2-pentenyl; 3-pentenyl; 4-pentenyl; 3 -methyl- 1-butenyl; 3- methyl-2-butenyl; 3-methyl-3-butenyl; 2-methyl- 1-butenyl; 2-methyl-2-butenyl; 2-methyl-3- butenyl; 2-ethyl-2-propenyl; 1 -methyl- 1-butenyl; l-methyl-2-butenyl; and l-methyl-3-butenyl. The teπn "alkynyl" generally refers to any structure or moiety having the unsaturation C≡C. Alkynyl groups include alkynyl groups substituted with: alkyl substituents or cycloalkyl substituents; or alkyl groups or cycloalkyl groups substituted with an alkynyl group. Examples of alkynyl groups include, but are not limited to: ethynyl; 1-propynyl; 2-propynyl; 1-butynyl; 2- butynyl; 3-butynyl; 1-pentynyl; 2-pentynyl; 3-pentynyl; 4-pentynyl; l-methyl-2-propynyl; 1- methyl-2-butynyl; l-methyl-3-butynyl; 2-methyl-3-butynyl; l,2-dimethyl-3-butynyl; and 2,2- dimethyl- 3- butynyl.
The term "pharmaceutically acceptable salts" as used herein 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, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. Pharmaceutically acceptable acid addition salts of the compounds of the invention include salts derived from inorganic acids such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorus, and the like, as well as the salts derived from organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. Such salts thus include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, phthalate, benzenesulfonate, toluenesulfonate, phenylacetate, citrate, lactate, maleate, tartrate, methanesulfonate, and the like. Also contemplated are the salts of amino acids such as arginate, gluconate, galacturonate, and the like; see, for example, Berge et al., "Pharmaceutical Salts," J. of Pharmaceutical Science, 1977; 66:1 19. The acid addition salts of the basic compounds are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt in the conventional manner. The free base form may be regenerated by contacting the salt form with a base, and isolating the free base in the conventional manner. The free base forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free base for purposes of the present invention. Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metal hydroxides, or of organic amines. Examples of metals used as cations are sodium, potassium, magnesium, calcium, and the like. Examples of suitable amines are N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine, and procaine; see, for example, Berge et al., supra., 1977. The base addition salts of acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. The free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in a conventional manner. The free acid forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acid for purposes of the present invention. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, the disclosure of which is hereby incorporated by reference.
In one embodiment, pharmaceutical preparations that include, as the active component any of the OCT3 inhibitors described above may be used to treat a subject. Any suitable route of administration may be employed for providing a patient with an effective dosage of compounds described above or pharmaceutically acceptable salts thereof. For example, oral, rectal, topical, parenteral, ocular, intracranial, pulmonary, nasal, and the like may be employed. Dosage forms may include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, oils, emulsions, liposomes, aerosols, and the like. In certain embodiments, it may be advantageous that the compositions described herein be administered orally. In other embodiments, it may be advantageous that the compositions described herein be administered parenterally. In yet other embodiments, it may be advantageous that the compositions described herein be administered locally, at the site of tissue injury.
The pharmaceutical compositions may include those compositions suitable for oral, rectal, topical, parenteral (including subcutaneous, intramuscular, and intravenous), ocular (ophthalmic), pulmonary (aerosol inhalation), or nasal administration, although the most suitable route in any given case will depend on the nature and severity of the conditions being treated and on the nature of the active ingredient. They may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy. For administration by inhalation, the compositions described herein are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or nebulizers. The compositions may also be delivered as powders which may be formulated and the powder composition may be inhaled with the aid of an insufflation powder inhaler device. Suitable topical formulations for use in the present embodiments may include transdermal devices, aerosols, creams, ointments, lotions, dusting powders, and the like.
In practical use, compositions can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). In preparing the compositions for oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, capsules and tablets, with the solid oral preparations being preferred over the liquid preparations. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques.
The pharmaceutical preparations may be manufactured in a manner which is itself known to one skilled in the art, for example, by means of conventional mixing, granulating, dragee- making, softgel encapsulation, dissolving, extracting, or lyophilizing processes. Thus, pharmaceutical preparations for oral use may be obtained by combining the active compounds with solid and semi- solid excipients and suitable preservatives, and/or co-antioxidants. Optionally, the resulting mixture may be ground and processed. The resulting mixture of granules may be used, after adding suitable auxiliaries, if desired or necessary, to obtain tablets, softgels, lozenges, capsules, or dragee cores.
Suitable excipients may be fillers such as saccharides (e.g., lactose, sucrose, or mannose), sugar alcohols (e.g., mannitol or sorbitol), cellulose preparations and/or calcium phosphates (e.g., tricalcium phosphate or calcium hydrogen phosphate). In addition binders may be used such as starch paste (e.g., maize or corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone). Disintegrating agents may be added (e.g., the above-mentioned starches) as well as carboxymethyl- starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof (e.g., sodium alginate). Auxiliaries are, above all, flow-regulating agents and lubricants (e.g., silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol, or PEG). Dragee cores are provided with suitable coatings, which, if desired, are resistant to gastric juices. Softgelatin capsules ("softgels") are provided with suitable coatings, which, typically, contain gelatin and/or suitable edible dye(s). Animal component-free and kosher gelatin capsules may be particularly suitable for the embodiments described herein for wide availability of usage and consumption. For this purpose, concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol (PEG) and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures, including dimethylsulfoxide (DMSO), tetrahydrofuran (THF), acetone, ethanol, or other suitable solvents and co-solvents. In order to produce coatings resistant to gastric juices, solutions of suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethyl-cellulose phthalate, may be used. Dye stuffs or pigments may be added to the tablets or dragee coatings or softgelatin capsules, for example, for identification or in order to characterize combinations of active compound doses, or to disguise the capsule contents for usage in clinical or other studies.
Other pharmaceutical preparations that may be used orally include push-fit capsules made of gelatin, as well as soft, thermally sealed capsules made of gelatin and a plasticizer such as glycerol or sorbitol. The push-fit capsules may contain the active compounds in the form of granules that may be mixed with fillers such as, for example, lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers and/or preservatives. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils such as rice bran oil or peanut oil or palm oil, or liquid paraffin. In some embodiments, stabilizers and preservatives may be added.
In some embodiments, pulmonary administration of a pharmaceutical composition may be desirable. Pulmonary administration may include, for example, inhalation of aerosolized or nebulized liquid or solid particles of the pharmaceutically active component dispersed in and surrounded by a gas.
Possible pharmaceutical preparations, which may be used rectally or vaginally, include, for example, suppositories, which include a combination of the active compounds with a suppository base. Suitable suppository bases are, for example, natural or synthetic triglycerides, or paraffin hydrocarbons. In addition, it is also possible to use gelatin rectal capsules that consist of a combination of the active compounds with a base. Possible base materials include, for example, liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
Suitable formulations for parenteral administration include, but are not limited to, aqueous solutions of the active compounds in water-soluble and/or water dispersible form, for example, water-soluble salts, esters, carbonates, phosphate esters or ethers, sulfates, glycoside ethers, together with spacers and/or linkers. Suspensions of the active compounds as appropriate oily injection suspensions may be administered, particularly suitable for intramuscular injection. Suitable lipophilic solvents, co-solvents (such as DMSO or ethanol), and/or vehicles including fatty oils, for example, rice bran oil or peanut oil and/or palm oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides, may be used. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension including, for example, sodium carboxymethyl cellulose, sorbitol, dextran, and/or cyclodextrins. Cyclodextrins (e.g., β- cyclodextrin) may be used specifically to increase the water solubility for parenteral injection of the compound. Liposomal formulations, in admixture with, for example, egg yolk phosphotidylcholine (E-PC), may be made for injection. Optionally, the suspension may contain stabilizers, for example, antioxidants such as BHT, and/or preservatives, such as benzyl alcohol.
The compositions of this invention can be administered in such oral dosage forms as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions, syrups, and emulsions. They may also be administered in intravenous (bolus or infusion), intraperitoneal, subcutaneous, or intramuscular form, all using dosage forms well known to those of ordinary skill in the pharmaceutical arts. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice. The dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired. A physician or veterinarian may determine and prescribe the effective amount of the drug required to prevent, counter, or arrest the progress or the development prostate cancer in a subject. The pharmaceutical compositions may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four or more times daily.
The pharmaceutical compositions described herein may further be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using transdermal skin patches. When administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
The compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as "pharmacologically inert carriers") suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices.
For instance, for oral administration in the form of a tablet or capsule, the pharmacologically active component may be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like; for oral administration in liquid form, the oral drug components can be combined with any oral, nontoxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents, and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
The compounds of the present invention may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine, or phosphatidylcholines.
Compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxide-polylysine substituted with palmitoyl residues. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacylates, and crosslinked or amphipathic block copolymers of hydrogels.
Gelatin capsules may contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract.
Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance. In general, water, a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene glycols are suitable carriers for parenteral solutions. Solutions for parenteral administration preferably contain a water-soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances. Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents. Also used are citric acid and its salts and sodium EDTA. In addition, parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propyl-paraben, and chlorobutanol.
Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, Mack Publishing Company, a standard reference text in this field. In an embodiment, the pharmaceutical composition may be administered to the patient systemically. The term systemic as used herein includes subcutaneous injection; intravenous, intramuscular, intraestemal injection; infusion; inhalation, transdermal administration, oral administration; and intra-operative instillation.
One systemic method involves an aerosol suspension of respirable particles comprising the active compound, which the subject inhales. The active compound would be absorbed into the bloodstream via the lungs, and subsequently contact the lacrimal glands in a pharmaceutically effective amount. The respirable particles may be liquid or solid, with a particle size sufficiently small to pass through the mouth and larynx upon inhalation; in general, particles ranging from about 1 to 10 microns, but more preferably 1-5 microns, in size are considered respirable.
Another method of systemically administering the active compounds involves administering a liquid/liquid suspension in the form of eye drops or eye wash or nasal drops of a liquid formulation, or a nasal spray of respirable particles that the subject inhales. Liquid pharmaceutical compositions of the active compound for producing a nasal spray or nasal or eye drops may be prepared by combining the active compound with a suitable vehicle, such as sterile pyrogen free water or sterile saline by techniques known to those skilled in the art.
The active compounds may also be systemically administered through absorption by the skin using transdermal patches or pads. The active compounds are absorbed into the bloodstream through the skin. Plasma concentration of the active compounds can be controlled by using patches containing different concentrations of active compounds.
Other methods of systemic administration of the active compound involves oral administration, in which pharmaceutical compositions containing active compounds are in the form of tablets, lozenges, aqueous or oily suspensions, viscous gels, chewable gums, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Additional means of systemic administration of the active compound to the subject may involve a suppository form of the active compound, such that a therapeutically effective amount of the compound reaches the eyes via systemic absorption and circulation. Further means of systemic administration of the active compound involve direct intraoperative instillation of a gel, cream, or liquid suspension form of a therapeutically effective amount of the active compound.
For topical application, the solution containing the active compound may contain a physiologically compatible vehicle, as those skilled in the art can select, using conventional criteria. The vehicles may be selected from the known pharmaceutical vehicles which include, but are not limited to, saline solution, water polyethers such as polyethylene glycol, polyvinyls such as polyvinyl alcohol and povidone, cellulose derivatives such as methylcellulose and hydroxypropyl methylcellulose, petroleum derivatives such as mineral oil and white petrolatum, animal fats such as lanolin, polymers of acrylic acid such as carboxypolymethylene gel, vegetable fats such as peanut oil and polysaccharides such as dextrans, and glycosaminoglycans such as sodium hyaluronate and salts such as sodium chloride and potassium chloride. For systemic administration such as injection and infusion, the pharmaceutical formulation is prepared in a sterile medium. The active ingredient, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Adjuvants such as local anaesthetics, preservatives and buffering agents can also be dissolved in the vehicle. The sterile injectable preparation may be a sterile injectable solution or suspension in a non-toxic acceptable diluent or solvent. Among the acceptable vehicles and solvents that may be employed are sterile water, saline solution, or Ringer's solution.
In practical use, the OCT3 inhibitor used may be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). In preparing the compositions for oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, capsules and tablets, with the solid oral preparations being preferred over the liquid preparations. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques.
Summary of Experimental Procedures
5-HTT mutant mice were generated according to the method of described in Bengel D, et al. (1998) "Altered brain serotonin (5-HT) homeostasis and locomotor insensitivity to MDMA ("ecstasy") in 5-HT transporter deficient mice." MoI Pharmacol 53:649-655. Male, 5-HTT+/+, 5 -HTT+/-, or 5-HTT -/- mice, weighing 25 to 30 g, were used for all experiments. RT-PCR was used to quantify OCT 1 and 3 mRNA according to the method of Chen J-J, et al. (2001) "Maintenance of serotonin in the intestinal mucosa and ganglia of mice that lack the high- affinity serotonin transporter: Abnormal intestinal motility and the expression of cation transporters." J Neurosci 21:6348-6361. Western blot analysis of OCT 1 and 3 protein expression was performed using commercial antibodies to OCT 1 and 3 (Alpha Diagnostics International) according to the method of Sata R, et al. (2005) "Functional analysis of organic cation transporter 3 expressed in human placenta." J Pharmacol Exp Ther 315:888-895. Tissue was assayed for protein content by the method of Bradford M-M (1976) "A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein- dye-binding. Anal Biochem 72:248-254. Immunocytochemical staining for OCT3 was performed using the same antibody as used for Western blot analyses. High-speed chronoamperometry was used according to established protocols (Daws L-C, Toney G-M (2007) "Voltammetric Methods to Study Kinetics and Mechanisms for Serotonin Clearance In Vivo." In, Electrochemical Methods in Neuroscience, for Methods and New Frontiers in Neuroscience, eds Michael A-C, Simon S-A, Nicolelis M-A-L. (CRC Press, Boca Raton, FL), pp 63-81) to measure clearance of 5-HT and histamine from extracellular fluid of hippocampus of anesthetized mice in vivo as well as the effect of locally applied D-22 on the clearance of these transmitters. All solutions were applied intrahippocampally by pressure-ejection and electrode placement verified histologically at the conclusion of each experiment. The TST was based on the method of Steru L, Chermat R, Thierry B, Simon P (1985) "The tail suspension test: A new method for screening antidepressants in mice." Psychopharmacology (Berl) 85:367-370.. Mice were injected intraperitoneally with either D-22 (1.0 μg/kg) or saline and then placed in an observation chamber for 30 min. Immediately following this period they were securely fastened by the distal end of the tail to a flat metallic surface and suspended in a visually isolated area (40 x 40 x 40 cm white box). The amount of time spent immobile, defined as the absence of limb movement, was recorded over a 6-min test session by a trained observer who remained blind to genotype and treatment. The mice underwent a second TST 60 min. following injection of D-22 or saline. The effects of genotype and drugs were analyzed using ANOVA followed by Bonferroni post-hoc tests. Data are presented as the mean and standard error of the mean (s.e.m.).
In this patent, certain U.S. patents, U.S. patent applications, and other materials (e.g., articles) have been incorporated by reference. The text of such U.S. patents, U.S. patent applications, and other materials is, however, only incorporated by reference to the extent that no conflict exists between such text and the other statements and drawings set forth herein. In the event of such conflict, then any such conflicting text in such incorporated by reference U.S. patents, U.S. patent applications, and other materials is specifically not incorporated by reference in this patent.
Further modifications and alternative embodiments of various aspects of the invention will be apparent to those skilled in the art in view of this description. Accordingly, this description is to be construed as illustrative only and is for the purpose of teaching those skilled in the art the general manner of carrying out the invention. It is to be understood that the forms of the invention shown and described herein are to be taken as examples of embodiments. Elements and materials may be substituted for those illustrated and described herein, parts and processes may be reversed, and certain features of the invention may be utilized independently, all as would be apparent to one skilled in the art after having the benefit of this description of the invention. Changes may be made in the elements described herein without departing from the spirit and scope of the invention as described in the following claims.

Claims

WHAT IS CLAIMED IS:
1. A method of inhibiting or reducing the effects of a mental disorder in a subject comprising administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising an active substance, wherein the active substance is an amino-catechol derivative.
2. The method of claim 1, wherein the amino-catechol derivative has the structure:
Figure imgf000026_0001
where R1 is hydrogen, alkyl, or -COOH; where R2 and I RE 3 are hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl; and where each R4 is independently hydrogen, alkyl, alkenyl, aryl or benzyl.
3. The method of claim 1, wherein the amino-catechol derivative has the structure:
Figure imgf000026_0002
where R1 is hydrogen, alkyl, or -COOH; where R2 is hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl; and where R4 is independently hydrogen, alkyl, alkenyl, aryl or benzyl.
4. The method of claim 1, wherein the amino-catechol derivative has the structure:
Figure imgf000027_0001
5. The method of claim 1, wherein the amino-catechol derivative has the structure:
Figure imgf000027_0002
6. The method of claim 1, wherein the amino-catechol derivative has the structure:
Figure imgf000027_0003
7. A method of inhibiting or reducing the effects of a mental disorder in a subject comprising administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising an active substance, wherein the active substance is a quinoline derivative.
8. The method of claim 7, wherein the quinoline derivative has the structure:
Figure imgf000027_0004
where each R is independently hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl; and R6 is hydrogen, alkyl, alkenyl, akynyl, or -CO2R5
9. The method of claim 7, wherein the quinoline derivative has the structure:
Figure imgf000028_0001
10. A method of inhibiting or reducing the effects of a mental disorder in a subject comprising administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising an active substance, wherein the active substance is a bis-quinoline derivative.
11. The method of claim 10, wherein the bis-quinoline derivative has the structure:
Figure imgf000028_0002
where each R is independently hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl.
12. The method of claim 10, wherein the bis-quinoline derivative has the structure:
Figure imgf000028_0003
where each R is independently hydrogen, alkyl, cycloalkyl, alkenyl, or akynyl.
13. The method of claim 10, wherein the bis-quinoline derivative has the structure:
Figure imgf000029_0001
14. The method of claim 10, wherein the bis-quinoline derivative has the structure:
Figure imgf000029_0002
15. The method of claim 10, wherein the bis-quinoline derivative has the structure:
Figure imgf000029_0003
16. The method of claim 10, wherein the bis-quinoline derivative has the structure:
Figure imgf000029_0004
17. The method of any one of claims 1-16, wherein the mental disorder is depression and the active-substance acts as an anti-depressant.
18. The method of any one of claims 1-16, wherein the mental disorder is an addiction to alcohol.
19. The method of any one of claims 1-16, wherein the mental disorder is an addiction to drugs.
20. The method of any one of claims 1-19, wherein the pharmaceutically acceptable formulation is administered by injection.
21. The method of any one of claims 1-19, wherein the pharmaceutically acceptable formulation is an oral dosage form.
22. A method of inhibiting or reducing the effects of a mental disorder in a subject comprising administering to a subject in need thereof an effective amount of a pharmaceutically acceptable formulation comprising an active substance, wherein the active substance is an OCT3 inhibitor.
PCT/US2009/042101 2008-04-29 2009-04-29 Therapeutics for treatment resistant mental disorders WO2009134877A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4879508P 2008-04-29 2008-04-29
US61/048,795 2008-04-29

Publications (2)

Publication Number Publication Date
WO2009134877A2 true WO2009134877A2 (en) 2009-11-05
WO2009134877A3 WO2009134877A3 (en) 2010-03-04

Family

ID=41255753

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/042101 WO2009134877A2 (en) 2008-04-29 2009-04-29 Therapeutics for treatment resistant mental disorders

Country Status (1)

Country Link
WO (1) WO2009134877A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014103801A1 (en) 2012-12-28 2014-07-03 株式会社新日本科学 Oct3 activity inhibitor containing imidazopyridine derivative as active component, and oct3 detection agent
WO2015002150A1 (en) 2013-07-03 2015-01-08 株式会社新日本科学 Novel compound, organic cation transporter 3 detection agent, and organic cation transporter 3 activity inhibitor
EP3428157A1 (en) * 2017-07-13 2019-01-16 Universitè Pierre Et Marie Curie - Paris 6 (Upmc) Selective organic cation transporters inhibitors for the treatment of depressive disorders

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6403645B2 (en) * 2000-03-16 2002-06-11 President And Fellows Of Harvard College Antidepressant effect of norepinephrine uptake 2 inhibitors and combined medications including them
US20070136828A1 (en) * 2004-03-09 2007-06-14 Biostation Inc. Methods of using molecules related to organic cation transporter 3 (oct3) for treating depression, anxiety neuroses, drug dependencies, and other similar mental disorders
WO2007100777A2 (en) * 2006-02-28 2007-09-07 The Mclean Hospital Corporation Methods for the treatment of adhd and related disorders

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6403645B2 (en) * 2000-03-16 2002-06-11 President And Fellows Of Harvard College Antidepressant effect of norepinephrine uptake 2 inhibitors and combined medications including them
US20070136828A1 (en) * 2004-03-09 2007-06-14 Biostation Inc. Methods of using molecules related to organic cation transporter 3 (oct3) for treating depression, anxiety neuroses, drug dependencies, and other similar mental disorders
WO2007100777A2 (en) * 2006-02-28 2007-09-07 The Mclean Hospital Corporation Methods for the treatment of adhd and related disorders

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
KITAICHI KIYOYUKI ET AL.: 'Behavioral changes following antisense oligonucleotide-induced reduction of organic cation transporter-3 in mice' NEUROSCIENCE LETTERS vol. 382, 2005, pages 195 - 200 *
NAKAYAMA HIRONAO ET AL.: 'The role of organic cation transporter-3 in methamphetamine disposition and its behavioral response in rats' BRAIN RESEARCH vol. 1184, 2007, pages 260 - 269 *
SCHILDKRAUT JOSEPH ET AL.: 'Toward a Rapidly Acting Antidepressant: The Normetanephrine and Extraneuronal Monoamine Transporter (Uptake 2) Hypothesis' AM J PSYCHIATRY vol. 161, no. 5, 2004, pages 909 - 911 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014103801A1 (en) 2012-12-28 2014-07-03 株式会社新日本科学 Oct3 activity inhibitor containing imidazopyridine derivative as active component, and oct3 detection agent
EP2939675A4 (en) * 2012-12-28 2016-09-14 Shin Nippon Biomedical Lab Ltd Oct3 activity inhibitor containing imidazopyridine derivative as active component, and oct3 detection agent
JPWO2014103801A1 (en) * 2012-12-28 2017-01-12 株式会社新日本科学 OCT3 activity inhibitor or OCT3 detector comprising an imidazopyridine derivative as an active ingredient
US10149840B2 (en) 2012-12-28 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. OCT3 activity inhibitor containing imidazopyridine derivative as active component, and OCT3 detection agent
WO2015002150A1 (en) 2013-07-03 2015-01-08 株式会社新日本科学 Novel compound, organic cation transporter 3 detection agent, and organic cation transporter 3 activity inhibitor
JPWO2015002150A1 (en) * 2013-07-03 2017-02-23 株式会社新日本科学 Novel compound, detection agent and activity inhibitor of organic cation transporter 3
US9745274B2 (en) 2013-07-03 2017-08-29 Shin Nippon Biomedical Laboratories, Ltd. Compound, organic cation transporter 3 detection agent, and organic cation transporter 3 activity inhibitor
EP3428157A1 (en) * 2017-07-13 2019-01-16 Universitè Pierre Et Marie Curie - Paris 6 (Upmc) Selective organic cation transporters inhibitors for the treatment of depressive disorders
WO2019012150A1 (en) * 2017-07-13 2019-01-17 Sorbonne Université Selective organic cation transporters inhibitors for the treatment of depressive disorders
US11414398B2 (en) 2017-07-13 2022-08-16 Sorbonne Université Selective organic cation transporters inhibitors for the treatment of depressive disorders

Also Published As

Publication number Publication date
WO2009134877A3 (en) 2010-03-04

Similar Documents

Publication Publication Date Title
Koivisto et al. Advances in TRP channel drug discovery: from target validation to clinical studies
Bonvini et al. Cough and airway disease: the role of ion channels
Chou et al. Differential effects of airway afferent nerve subtypes on cough and respiration in anesthetized guinea pigs
Li et al. Oral dimethyl fumarate reduces peripheral neuropathic pain in rodents via NFE2L2 antioxidant signaling
Lin et al. Activations of TRPA1 and P2X receptors are important in ROS-mediated stimulation of capsaicin-sensitive lung vagal afferents by cigarette smoke in rats
Kenyon et al. Self-assembling nanoparticles containing dexamethasone as a novel therapy in allergic airways inflammation
Napimoga et al. 15d-prostaglandin J2 inhibits inflammatory hypernociception: involvement of peripheral opioid receptor
Pena-Dos-Santos et al. Activation of peripheral κ/δ opioid receptors mediates 15-deoxy-Δ12, 14-prostaglandin J2 induced-antinociception in rat temporomandibular joint
Craft et al. Sex differences in cocaine-and nicotine-induced antinociception in the rat
Lynch III et al. ABT-594 (a nicotinic acetylcholine agonist): anti-allodynia in a rat chemotherapy-induced pain model
Hughes Acute chloroquine poisoning: A comprehensive experimental toxicology assessment of the role of diazepam
JP2023516284A (en) Compositions and therapeutic uses of cannabidiol
EP1986619A2 (en) Use of phosphatases to treat tumors overexpressing n-cor
Li et al. Toxicity mechanism of nanomaterials: Focus on endoplasmic reticulum stress
Torabi et al. Adolescent nicotine challenge promotes the future vulnerability to opioid addiction: Involvement of lateral paragigantocellularis neurons
Zhang et al. Role of µ, κ, and δ opioid receptors in tibial inhibition of bladder overactivity in cats
WO2009134877A2 (en) Therapeutics for treatment resistant mental disorders
Jiang et al. Spinal neuronal NOS signaling contributes to morphine cardioprotection in ischemia reperfusion injury in rats
Zhang et al. Polyethylenimine-modified mesoporous silica nanoparticles induce a survival mechanism in vascular endothelial cells via microvesicle-mediated autophagosome release
Wex et al. The long‐acting β2‐adrenoceptor agonist olodaterol attenuates pulmonary inflammation
Kyriakou et al. Efficacy of cannabinoids against glioblastoma multiforme: A systematic review
US20200405657A1 (en) Treatment of pain using allosteric modulator of trpv1
KR20220025849A (en) Carbocyanine compounds for targeting of mitochondria and eradication of cancer stem cells
Lin et al. Perivagal antagonist treatment in rats selectively blocks the reflex and afferent responses of vagal lung C fibers to intravenous agonists
Reynolds et al. Adenosine induces a cholinergic tracheal reflex contraction in guinea pigs in vivo via an adenosine A1 receptor-dependent mechanism

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: 09739679

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09739679

Country of ref document: EP

Kind code of ref document: A2