WO2010060070A2 - Modulateurs à base d'indolinone du récepteur de la dopamine - Google Patents

Modulateurs à base d'indolinone du récepteur de la dopamine Download PDF

Info

Publication number
WO2010060070A2
WO2010060070A2 PCT/US2009/065637 US2009065637W WO2010060070A2 WO 2010060070 A2 WO2010060070 A2 WO 2010060070A2 US 2009065637 W US2009065637 W US 2009065637W WO 2010060070 A2 WO2010060070 A2 WO 2010060070A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
recited
deuterium
group
acid
Prior art date
Application number
PCT/US2009/065637
Other languages
English (en)
Other versions
WO2010060070A3 (fr
Inventor
Thomas G. Gant
Sepehr Sarshar
Original Assignee
Auspex Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Auspex Pharmaceuticals, Inc. filed Critical Auspex Pharmaceuticals, Inc.
Publication of WO2010060070A2 publication Critical patent/WO2010060070A2/fr
Publication of WO2010060070A3 publication Critical patent/WO2010060070A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/32Oxygen atoms
    • C07D209/34Oxygen atoms in position 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system

Definitions

  • indolinone compounds and compositions and their application as pharmaceuticals for the treatment of disorders are also provided for the treatment of disorders such as restless leg syndrome and Parkinson's disease.
  • Ropinirole (Adartrel®, ReQuip®, Requip XL®, Ropark®, SK&F 101468, SK&F 101468-A, CAS # 91374-20-8), 4-(2-(dipropylamino)ethyl)indolin- 2-one, is a dopamine receptor agonist.
  • Ropinirole is commonly prescribed for the treatment of restless leg syndrome and Parkinson's disease.
  • Ropinirole is subject to CYP1A2- and CYP3A-mediated metabolic oxidation of the three methylene groups alpha to the amine nitrogen to give N- despropyl and des-diisopropylamine metabolites, as well as hydroxylation of the 7- position of the indolinone ring. Bloomer et al., Drug Metab. Disp., 1997, 25(7), 840-844; Kaye et al., CHn. Pharmacokinet, 2000, 39(4), 243-254; Matheson et al., Drugs, 2000, 60(1), 115-137; Reavill et al., /. Pharm.
  • Adverse effects associated with ropinirole include dizziness, drowsiness, fatigue, headache, heartburn, vomiting, constipation, urinary frequency, mouth dryness, and decreased libido.
  • the animal body expresses various enzymes, such as the cytochrome P 450 enzymes (CYPs), esterases, proteases, reductases, dehydrogenases, and monoamine oxidases, to react with and convert these foreign substances to more polar intermediates or metabolites for renal excretion.
  • CYPs cytochrome P 450 enzymes
  • esterases proteases
  • reductases reductases
  • dehydrogenases dehydrogenases
  • monoamine oxidases monoamine oxidases
  • Such metabolic reactions frequently involve the oxidation of a carbon-hydrogen (C-H) bond to either a carbon-oxygen (C-O) or a carbon-carbon (C-C) ⁇ -bond.
  • C-H carbon-hydrogen
  • C-O carbon-oxygen
  • C-C carbon-carbon
  • the resultant metabolites may be stable or unstable under physiological conditions, and can have substantially different pharmacokinetic, pharmacodynamic, and acute and long-term
  • the transition state in a reaction is a short lived state along the reaction pathway during which the original bonds have stretched to their limit.
  • the activation energy E 301 for a reaction is the energy required to reach the transition state of that reaction. Once the transition state is reached, the molecules can either revert to the original reactants, or form new bonds giving rise to reaction products.
  • a catalyst facilitates a reaction process by lowering the activation energy leading to a transition state. Enzymes are examples of biological catalysts.
  • Carbon-hydrogen bond strength is directly proportional to the absolute value of the ground- state vibrational energy of the bond. This vibrational energy depends on the mass of the atoms that form the bond, and increases as the mass of one or both of the atoms making the bond increases. Since deuterium (D) has twice the mass of protium ( 1 H), a C-D bond is stronger than the corresponding C- 1 H bond. If a C- 1 H bond is broken during a rate-determining step in a chemical reaction (i.e. the step with the highest transition state energy), then substituting a deuterium for that protium will cause a decrease in the reaction rate. This phenomenon is known as the Deuterium Kinetic Isotope Effect (DKIE).
  • DKIE Deuterium Kinetic Isotope Effect
  • the magnitude of the DKIE can be expressed as the ratio between the rates of a given reaction in which a C- 1 H bond is broken, and the same reaction where deuterium is substituted for protium.
  • the DKIE can range from about 1 (no isotope effect) to very large numbers, such as 50 or more. Substitution of tritium for hydrogen results in yet a stronger bond than deuterium and gives numerically larger isotope effects [0009]
  • Deuterium ( 2 H or D) is a stable and non-radioactive isotope of hydrogen which has approximately twice the mass of protium ( 1 H), the most common isotope of hydrogen.
  • Deuterium oxide (D 2 O or "heavy water”) looks and tastes like H 2 O, but has different physical properties.
  • the DKIE was used to decrease the hepatotoxicity of halothane, presumably by limiting the production of reactive species such as trifluoroacetyl chloride.
  • this method may not be applicable to all drug classes.
  • deuterium incorporation can lead to metabolic switching.
  • Metabolic switching occurs when xenogens, sequestered by Phase I enzymes, bind transiently and re-bind in a variety of conformations prior to the chemical reaction (e.g., oxidation). Metabolic switching is enabled by the relatively vast size of binding pockets in many Phase I enzymes and the promiscuous nature of many metabolic reactions. Metabolic switching can lead to different proportions of known metabolites as well as altogether new metabolites. This new metabolic profile may impart more or less toxicity. Such pitfalls are non- obvious and are not predictable a priori for any drug class.
  • Ropinirole is a dopamine receptor agonist.
  • the carbon-hydrogen bonds of ropinirole contain a naturally occurring distribution of hydrogen isotopes, namely 1 H or protium (about 99.9844%), 2 H or deuterium (about 0.0156%), and 3 H or tritium (in the range between about 0.5 and 67 tritium atoms per 10 18 protium atoms).
  • Increased levels of deuterium incorporation may produce a detectable Deuterium Kinetic Isotope Effect (DKIE) that could effect the pharmacokinetic, pharmacologic and/or toxicologic profiles of such ropinirole in comparison with the compound having naturally occurring levels of deuterium.
  • DKIE Deuterium Kinetic Isotope Effect
  • a medicine with a longer half-life may result in greater efficacy and cost savings.
  • Various deuteration patterns can be used to (a) reduce or eliminate unwanted metabolites, (b) increase the half-life of the parent drug, (c) decrease the number of doses needed to achieve a desired effect, (d) decrease the amount of a dose needed to achieve a desired effect, (e) increase the formation of active metabolites, if any are formed, (f) decrease the production of deleterious metabolites in specific tissues, and/or (g) create a more effective drug and/or a safer drug for polypharmacy, whether the polypharmacy be intentional or not.
  • the deuteration approach has the strong potential to slow the metabolism of ropinirole and attenuate interpatient variability.
  • Novel compounds and pharmaceutical compositions certain of which have been found to modulate dopamine receptor have been discovered, together with methods of synthesizing and using the compounds, including methods for the treatment of dopamine receptor-mediated disorders in a patient by administering the compounds.
  • R 1 -R 24 are independently selected from the group consisting of hydrogen and deuterium; and at least one of R 1 -R 24 is deuterium.
  • Certain compounds disclosed herein may possess useful dopamine receptor modulating activity, and may be used in the treatment or prophylaxis of a disorder in which dopamine receptor plays an active role.
  • certain embodiments also provide pharmaceutical compositions comprising one or more compounds disclosed herein together with a pharmaceutically acceptable carrier, as well as methods of making and using the compounds and compositions.
  • Certain embodiments provide methods for modulating dopamine receptor.
  • Other embodiments provide methods for treating a dopamine receptor-mediated disorder in a patient in need of such treatment, comprising administering to said patient a therapeutically effective amount of a compound or composition according to the present invention.
  • certain compounds disclosed herein for use in the manufacture of a medicament for the prevention or treatment of a disorder ameliorated by the modulation of dopamine receptor.
  • the compounds as disclosed herein may also contain less prevalent isotopes for other elements, including, but not limited to, 13 C or 14 C for carbon, 33 S,
  • the compound disclosed herein may expose a patient to a maximum of about 0.000005% D 2 O or about 0.00001% DHO, assuming that all of the C-D bonds in the compound as disclosed herein are metabolized and released as D 2 O or DHO. In certain embodiments, the levels of
  • the deuterium-enriched compound disclosed herein should not cause any additional toxicity due to the formation of
  • the deuterated compounds disclosed herein maintain the beneficial aspects of the corresponding non-isotopically enriched molecules while substantially increasing the maximum tolerated dose, decreasing toxicity, increasing the half-life (Ty 2 ), lowering the maximum plasma concentration
  • C max of the minimum efficacious dose (MED), lowering the efficacious dose and thus decreasing the non-mechanism-related toxicity, and/or lowering the probability of drug-drug interactions.
  • Ri-R 2 are deuterium, then at least one of R 3 -
  • R 24 is deuterium
  • Ri-R 2 and R 4 -R 6 are deuterium, then at least one of R 3 and R 7 -R 24 is deuterium.
  • R 3 and R 7 -R 24 is deuterium.
  • R 7 -R 10 are deuterium, then at least one of Ri - R 6 and Rn-R 24 is deuterium.
  • Rn-Rn are deuterium, then at least one of R 1 - Rio and R] 8 -R 24 is deuterium.
  • Rn-R 24 are deuterium, then at least one of R 1 - Rio is deuterium.
  • Ri-R 24 is not deuterium.
  • All publications and references cited herein are expressly incorporated herein by reference in their entirety. However, with respect to any similar or identical terms found in both the incorporated publications or references and those explicitly put forth or defined in this document, then those terms definitions or meanings explicitly put forth in this document shall control in all respects. [0029] As used herein, the terms below have the meanings indicated. [0030] The singular forms "a,” “an,” and “the” may refer to plural articles unless specifically stated otherwise.
  • deuterium enrichment refers to the percentage of incorporation of deuterium at a given position in a molecule in the place of hydrogen. For example, deuterium enrichment of 1% at a given position means that 1% of molecules in a given sample contain deuterium at the specified position. Because the naturally occurring distribution of deuterium is about 0.0156%, deuterium enrichment at any position in a compound synthesized using non- enriched starting materials is about 0.0156%. The deuterium enrichment can be determined using conventional analytical methods known to one of ordinary skill in the art, including mass spectrometry and nuclear magnetic resonance spectroscopy.
  • deuterium when used to describe a given position in a molecule such as R 1 -R 24 or the symbol "D", when used to represent a given position in a drawing of a molecular structure, means that the specified position is enriched with deuterium above the naturally occurring distribution of deuterium.
  • deuterium enrichment is no less than about 1%, in another no less than about 5%, in another no less than about 10%, in another no less than about 20%, in another no less than about 50%, in another no less than about 70%, in another no less than about 80%, in another no less than about 90%, or in another no less than about 98% of deuterium at the specified position.
  • isotopic enrichment refers to the percentage of incorporation of a less prevalent isotope of an element at a given position in a molecule in the place of the more prevalent isotope of the element.
  • non-isotopically enriched refers to a molecule in which the percentages of the various isotopes are substantially the same as the naturally occurring percentages.
  • Asymmetric centers exist in the compounds disclosed herein. These centers are designated by the symbols “R” or “S,” depending on the configuration of substituents around the chiral carbon atom. It should be understood that the invention encompasses all stereochemical isomeric forms, including diastereomeric, enantiomeric, and epimeric forms, as well as d-isomers and 1-isomers, and mixtures thereof.
  • Individual stereoisomers of compounds can be prepared synthetically from commercially available starting materials which contain chiral centers or by preparation of mixtures of enantiomeric products followed by separation such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, direct separation of enantiomers on chiral chromatographic columns, or any other appropriate method known in the art.
  • Starting compounds of particular stereochemistry are either commercially available or can be made and resolved by techniques known in the art.
  • the compounds disclosed herein may exist as geometric isomers.
  • the present invention includes all cis, trans, syn, anti,
  • bonds refers to a covalent linkage between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure. A bond may be single, double, or triple unless otherwise specified. A dashed line between two atoms in a drawing of a molecule indicates that an additional bond may be present or absent at that position.
  • disorder as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disease,” “syndrome,” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms.
  • condition as in medical condition
  • the terms “treat,” “treating,” and “treatment” are meant to include alleviating or abrogating a disorder or one or more of the symptoms associated with a disorder; or alleviating or eradicating the cause(s) of the disorder itself.
  • treatment of a disorder is intended to include prevention.
  • prevent refers to a method of delaying or precluding the onset of a disorder; and/or its attendant symptoms, barring a subject from acquiring a disorder or reducing a subject's risk of acquiring a disorder.
  • therapeutically effective amount refers to the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disorder being treated.
  • therapeuticically effective amount also refers to the amount of a compound that is sufficient to elicit the biological or medical response of a cell, tissue, system, animal, or human that is being sought by a researcher, veterinarian, medical doctor, or clinician.
  • subject refers to an animal, including, but not limited to, a primate (e.g., human, monkey, chimpanzee, gorilla, and the like), rodents (e.g., rats, mice, gerbils, hamsters, ferrets, and the like), lagomorphs, swine (e.g., pig, miniature pig), equine, canine, feline, and the like.
  • a primate e.g., human, monkey, chimpanzee, gorilla, and the like
  • rodents e.g., rats, mice, gerbils, hamsters, ferrets, and the like
  • lagomorphs e.g., pig, miniature pig
  • swine e.g., pig, miniature pig
  • equine canine
  • feline feline
  • Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient.
  • administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the disorders described herein.
  • dopamine receptor refers to a subtype of receptor that binds dopamine.
  • Dopamine is a hormone and neurotransmitter occurring in a wide variety of animals. Five types of dopamine receptors are known - Dl, D2, D3, D4 and D5. Dopamine is produced in several areas of the brain, including the substantia nigra and the ventral tegmental area. Dopamine is also a neurohormone released by the hypothalamus. Its main function as a hormone is to inhibit the release of prolactin from the anterior lobe of the pituitary.
  • Dopamine receptors have key roles in many processes, including control of motivation, learning, and fine motor movement, as well as modulation of neuroendocrine signaling. Abnormal dopamine receptor signaling and dopaminergic nerve function is implicated in several neuropsychiatric disorders.
  • dopamine receptor-mediated disorder refers to a disorder that is characterized by abnormal dopamine receptor activity.
  • a dopamine receptor-mediated disorder may be completely or partially mediated by modulating dopamine receptor.
  • a dopamine receptor-mediated disorder is one in which modulation of dopamine receptor results in some effect on the underlying disorder e.g., administration of a dopamine receptor modulator results in some improvement in at least some of the patients being treated.
  • dopamine receptor modulator refers to the ability of a compound disclosed herein to alter the function of dopamine receptors.
  • a modulator may activate the activity of a dopamine receptor, may activate or inhibit the activity of a dopamine receptor depending on the concentration of the compound exposed to the dopamine receptor, or may inhibit the activity of a dopamine receptor. Such activation or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types.
  • modulate or “modulation” also refers to altering the function of a dopamine receptor by increasing or decreasing the probability that a complex forms between a dopamine receptor and a natural binding partner.
  • a modulator may increase the probability that such a complex forms between the dopamine receptor and the natural binding partner, may increase or decrease the probability that a complex forms between the dopamine receptor and the natural binding partner depending on the concentration of the compound exposed to the dopamine receptor, and or may decrease the probability that a complex forms between the dopamine receptor and the natural binding partner.
  • modulation of the dopamine receptor may be assessed using the method described in Reavill et al., J. Pharm. Pharmacol., 2000, 52(9), 1129-1135.
  • modulating dopamine receptor activity refers to altering the activity of dopamine receptors by administering a dopamine receptor modulator.
  • terapéuticaally acceptable refers to those compounds (or salts, prodrugs, tautomers, zwitterionic forms, etc.) which are suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, immunogenecity, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
  • pharmaceutically acceptable carrier refers to a pharmaceutically- acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material.
  • Each component must be “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation. It must also be suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenecity, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • active ingredient refers to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients or carriers, to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder.
  • drug refers to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder.
  • release controlling excipient refers to an excipient whose primary function is to modify the duration or place of release of the active substance from a dosage form as compared with a conventional immediate release dosage form.
  • nonrelease controlling excipient refers to an excipient whose primary function do not include modifying the duration or place of release of the active substance from a dosage form as compared with a conventional immediate release dosage form.
  • prodrug refers to a compound functional derivative of the compound as disclosed herein and is readily convertible into the parent compound in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound. A prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. See Harper, Progress in Drug Research 1962, 4, 221-294; Morozowich et al. in "Design of Biopharmaceutical Properties through Prodrugs and Analogs," Roche Ed., APHA Acad. Pharm. Sci.
  • the compounds disclosed herein can exist as therapeutically acceptable salts.
  • the term "therapeutically acceptable salt,” as used herein, represents salts or zwitterionic forms of the compounds disclosed herein which are therapeutically acceptable as defined herein.
  • the salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound with a suitable acid or base.
  • Therapeutically acceptable salts include acid and basic addition salts.
  • “Handbook of Pharmaceutical Salts, Properties, and Use” Stah and Wermuth, Ed. ;( Wiley- VCH and VHCA, Zurich, 2002) and Berge et al., /. Pharm. Sci. 1977, 66, 1-19.
  • Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, (+)- camphoric acid, camphorsulfonic acid, (+)-(lS)-camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane- 1 ,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, gluco
  • Suitable bases for use in the preparation of pharmaceutically acceptable salts including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol, diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2- (diethylamino)-ethanol, ethanolamine, ethylamine, ethylenediamine, isopropylamine, N-methyl-glucamine, hydrabamine, lH-imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine, l-
  • compositions which comprise one or more of certain compounds disclosed herein, or one or more pharmaceutically acceptable salts, prodrugs, or solvates thereof, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients.
  • pharmaceutical compositions which comprise one or more of certain compounds disclosed herein, or one or more pharmaceutically acceptable salts, prodrugs, or solvates thereof, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients.
  • Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g. , in Remington's Pharmaceutical Sciences.
  • compositions disclosed herein may be manufactured in any manner known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • the pharmaceutical compositions may also be formulated as a modified release dosage form, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms.
  • dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Deliver Technology, Rathbone et al., Eds., Drugs and the Pharmaceutical Science, Marcel Dekker, Inc.: New York, NY, 2002; Vol. 126).
  • compositions include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary), intraperitoneal, transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Typically, these methods include the step of bringing into association a compound of the subject invention or a pharmaceutically salt, prodrug, or solvate thereof ("active ingredient”) with the carrier which constitutes one or more accessory ingredients.
  • active ingredient a compound of the subject invention or a pharmaceutically salt, prodrug, or solvate thereof
  • the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • Formulations of the compounds disclosed herein suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a nonaqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • compositions which can be used orally include tablets, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • Dragee cores are provided with suitable coatings.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen-free water, immediately prior to use.
  • sterile liquid carrier for example, saline or sterile pyrogen-free water
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Formulations for parenteral administration include aqueous and nonaqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner.
  • Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth.
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides.
  • Certain compounds disclosed herein may be administered topically, that is by non-systemic administration. This includes the application of a compound disclosed herein externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream.
  • systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration.
  • Formulations suitable for topical administration include liquid or semi- liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose.
  • compounds may be delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray.
  • Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the compounds according to the invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
  • Preferred unit dosage formulations are those containing an effective dose, as herein below recited, or an appropriate fraction thereof, of the active ingredient.
  • Compounds may be administered orally or via injection at a dose of from 0.1 to 500 mg/kg per day.
  • the dose range for adult humans is generally from 5 mg to 2 g/day.
  • Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of one or more compounds which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the compounds can be administered in various modes, e.g. orally, topically, or by injection.
  • the precise amount of compound administered to a patient will be the responsibility of the attendant physician.
  • the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diets, time of administration, route of administration, rate of excretion, drug combination, the precise disorder being treated, and the severity of the disorder being treated. Also, the route of administration may vary depending on the disorder and its severity.
  • the administration of the compounds may be administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disorder.
  • the administration of the compounds may be given continuously or temporarily suspended for a certain length of time (i.e., a "drug holiday").
  • a maintenance dose is administered if necessary.
  • the dosage or the frequency of administration, or both can be reduced, as a function of the symptoms, to a level at which the improved disorder is retained. Patients can, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.
  • Dopamine receptor-mediated disorders include, but are not limited to, restless leg syndrome and Parkinson's disease, and/or any disorder which can lessened, alleviated, or prevented by administering a dopamine receptor modulator.
  • a method of treating a dopamine receptor- mediated disorder comprises administering to the subject a therapeutically effective amount of a compound of as disclosed herein, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, so as to affect: (1) decreased inter- individual variation in plasma levels of the compound or a metabolite thereof; (2) increased average plasma levels of the compound or decreased average plasma levels of at least one metabolite of the compound per dosage unit; (3) decreased inhibition of, and/or metabolism by at least one cytochrome P 450 or monoamine oxidase isoform in the subject; (4) decreased metabolism via at least one polymorphically-expressed cytochrome P 450 isoform in the subject; (5) at least one statistically-significantly improved disorder-control and/or disorder-eradication endpoint; (6) an improved clinical effect during the treatment of the disorder, (7) prevention of recurrence, or delay of decline or appearance, of abnormal alimentary or hepatic parameters as the primary clinical benefit, or (8) reduction
  • inter-individual variation in plasma levels of the compounds as disclosed herein, or metabolites thereof is decreased; average plasma levels of the compound as disclosed herein are increased; average plasma levels of a metabolite of the compound as disclosed herein are decreased; inhibition of a cytochrome P 450 or monoamine oxidase isoform by a compound as disclosed herein is decreased; or metabolism of the compound as disclosed herein by at least one polymorphic ally-expressed cytochrome P 450 isoform is decreased; by greater than about 5%, greater than about 10%, greater than about 20%, greater than about 30%, greater than about 40%, or by greater than about 50% as compared to the corresponding non-isotopically enriched compound.
  • Plasma levels of the compound as disclosed herein, or metabolites thereof may be measured using the methods described by Li et al. Rapid Communications in Mass Spectrometry 2005, 19, 1943-1950, Hughes et al, Xenobiotica 1992, 22(7), 859-69, Varma et al, Journal of Pharmaceutical and Biomedical Analysis 2004, 36(3), 669-674, Massoud et al, Journal of Chromatography, B: Biomedical Sciences and Applications 1999, 734(1), 163- 167, Kim et al, Journal of Pharmaceutical and Biomedical Analysis 2003, 31(2), 341-349, and Lindeke et al, Acta Pharmaceutica Suecica 1981, 18(1), 25-34.
  • Examples of cytochrome P 450 isoforms in a mammalian subject include, but are not limited to, CYPlAl, CYP1A2, CYPlBl, CYP2A6, CYP2A13, CYP2B6, CYP2C8, CYP2C9, CYP2C18, CYP2C19, CYP2D6, CYP2E1, CYP2G1, CYP2J2, CYP2R1, CYP2S1, CYP3A4, CYP3A5, CYP3A5P1, CYP3A5P2, CYP3A7, CYP4A11, CYP4B1, CYP4F2, CYP4F3, CYP4F8, CYP4F11, CYP4F12, CYP4X1, CYP4Z1, CYP5A1, CYP7A1, CYP7B1, CYP8A1, CYP8
  • the inhibition of the cytochrome P 450 isoform is measured by the method of Ko et al. (British Journal of Clinical Pharmacology, 2000, 49, 343-351).
  • the inhibition of the MAO A isoform is measured by the method of Weyler et al. (/. Biol Chem. 1985, 260, 13199-13207).
  • the inhibition of the MA0 B isoform is measured by the method of Uebelhack et al. (Pharmacopsychiatry, 1998, 31, 187- 192).
  • Examples of polymorphically-expressed cytochrome P 450 isoforms in a mammalian subject include, but are not limited to, CYP2C8, CYP2C9, CYP2C19, and CYP2D6.
  • liver microsomes cytochrome P 450 isoforms
  • monoamine oxidase isoforms are measured by the methods described herein.
  • improved disorder-control and/or disorder-eradication endpoints, or improved clinical effects include, but are not limited to, change from baseline in Japanese Unified Parkinson Disease Rating Scale (UPDRS) part III total score, improved International Restless Leg Syndrome Study Group Rating Scale (IRLS) scores, reduced number of leg movements per min, reduced average number of night awakenings, reduced periodic leg movements during sleep (PLMS), improved Clinical Global Impression scale scores, improved Medical Outcomes Study Sleep Scale scores, reduced periodic limb movements associated with arousal (PLMAI), change from baseline in sleep latency, and improved Unified Parkinson's Disease Rating Scale (UPDRS) motor scores.
  • UPDS Unified Parkinson Disease Rating Scale
  • IRLS International Restless Leg Syndrome Study Group Rating Scale
  • PLMS reduced number of leg movements per min
  • PLMS reduced average number of night awakenings
  • PLMS reduced periodic leg movements
  • hepatobiliary function endpoints include, but are not limited to, alanine aminotransferase ("ALT”), serum glutamic-pyruvic transaminase (“SGPT”), aspartate aminotransferase (“AST” or “SGOT”), ALT/AST ratios, serum aldolase, alkaline phosphatase (“ALP”), ammonia levels, bilirubin, gamma-glutamyl transpeptidase ("GGTP,” “ ⁇ -GTP,” or “GGT”), leucine aminopeptidase (“LAP”), liver biopsy, liver ultrasonography, liver nuclear scan, 5'- nucleotidase, and blood protein. Hepatobiliary endpoints are compared to the stated normal levels as given in "Diagnostic and Laboratory Test Reference", 4 th edition, Mosby, 1999. These assays are run by accredited laboratories according to standard protocol.
  • certain compounds and formulations disclosed herein may also be useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats.
  • the compounds disclosed herein may also be combined or used in combination with other agents useful in the treatment of dopamine receptor- mediated disorders.
  • the therapeutic effectiveness of one of the compounds described herein may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may only have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced).
  • Such other agents, adjuvants, or drugs may be administered, by a route and in an amount commonly used therefor, simultaneously or sequentially with a compound as disclosed herein.
  • a pharmaceutical composition containing such other drugs in addition to the compound disclosed herein may be utilized, but is not required.
  • the compounds disclosed herein can be combined with one or more dopamine agonists, antiepileptics, anticholinergics, anti-spasmodics, and L-dopa derivatives.
  • the compounds disclosed herein can be combined with a dopamine agonists known in the art, including, but not limited to, A-412,997, apomorphine, bromocriptine, cabergoline, dihydrexidine, dihydroergocryptine mesylate, fenoldopam, lisuride, pergolide, piribedil, pramipexole, propylnorapomorphine, quinpirole, ropinirole, rotigotine, SKF 38393, and SKF 82958.
  • a dopamine agonists known in the art, including, but not limited to, A-412,997, apomorphine, bromocriptine, cabergoline, dihydrexidine, dihydroergocryptine mesylate, fenoldopam, lisuride, pergolide, piribedil, pramipexole, propylnorapomorphine, quinpirole, ropini
  • the compounds disclosed herein can be combined with an antiepileptics known in the art, including, but not limited to, methylphenobarbital, phenobarbital, primidone, barbexaclone, metharbital, ethotoin, phenytoin, amino(diphenylhydantoin) valeric acid, mephenytoin, fosphenytoin, paramethadione, trimethadione, ethadione, ethosuximide, phensuximide, mesuximide, clonazepam, carbamazepine, oxcarbazepine, rufinamide, valproic acid, valpromide, aminobutyric acid, vigabatrin, progabide, tiagabine, sultiame, phenacemide, lamotrigine, felbamate, topiramate, gabapentin, pheneturide, le
  • the compounds provided herein can be combined with one or more anticholinergics known in the art, including, but not limited to, oxyphencyclimine, camylofin, mebeverine, trimebutine, rociverine, dicycloverine, dihexyverine, difemerine, piperidolate, benzilone, glycopyrronium, oxyphenonium, penthienate, propantheline, otilonium bromide, methantheline, tridihexethyl, isopropamide, hexocyclium, poldine, mepenzolate, bevonium, pipenzolate, biphemanil, (2-benzhydryloxyethyl)diethyl-methylammonium iodide, tiemonium iodide, prifinium bromide, timepidium bromide, and fenpiverinium.
  • anticholinergics known in the art, including, but not limited to, oxyphencyclimine,
  • the compounds disclosed herein can be combined with one or more anti-spasmodics known in the art, including, but not limited to, darifenacin, emepronium, flavoxate, meladrazine, oxybutynin, propiverine, solifenacin, terodiline, tolterodine, trospium, dimethylaminopropionylphenothiazine, nicofetamide, tiropramide, papaverine, drotaverine, and moxaverine.
  • anti-spasmodics known in the art, including, but not limited to, darifenacin, emepronium, flavoxate, meladrazine, oxybutynin, propiverine, solifenacin, terodiline, tolterodine, trospium, dimethylaminopropionylphenothiazine, nicofetamide, tiropramide, papaverine, d
  • the compounds disclosed herein can be combined with one or more L-dopa derivatives known in the art, including, but not limited to droxidopa, levodopa, melevodopa, and etilevodopa.
  • the compounds disclosed herein can also be administered in combination with other classes of compounds, including, but not limited to, norepinephrine reuptake inhibitors (NRIs) such as atomoxetine; dopamine reuptake inhibitors (DARIs), such as methylphenidate; serotonin-norepinephrine reuptake inhibitors (SNRIs), such as milnacipran; sedatives, such as diazepham; norepinephrine-dopamine reuptake inhibitor (NDRIs), such as bupropion; serotonin-norepinephrine-dopamine-reuptake-inhibitors (SNDRIs), such as venlafaxine; monoamine oxidase inhibitors, such as selegiline; hypothalamic phospholipids; endothelin converting enzyme (ECE) inhibitors, such as phosphoramidon; opioids, such as tramadol; thromboxane
  • squalene synthetase inhibitors include fibrates; bile acid sequestrants, such as questran; niacin; anti- atherosclerotic agents, such as ACAT inhibitors; MTP Inhibitors; calcium channel blockers, such as amlodipine besylate; potassium channel activators; alpha- muscarinic agents; beta-muscarinic agents, such as carvedilol and metoprolol; antiarrhythmic agents; diuretics, such as chlorothiazide, hydrochiorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichioromethiazide, polythiazide, benzothlazide, ethacrynic acid,
  • metformin glucosidase inhibitors
  • glucosidase inhibitors e.g., acarbose
  • insulins meglitinides (e.g., repaglinide)
  • meglitinides e.g., repaglinide
  • sulfonylureas e.g., glimepiride, glyburide, and glipizide
  • thiozolidinediones e.g.
  • certain embodiments provide methods for treating dopamine receptor-mediated disorders in a human or animal subject in need of such treatment comprising administering to said subject an amount of a compound disclosed herein effective to reduce or prevent said disorder in the subject, in combination with at least one additional agent for the treatment of said disorder that is known in the art.
  • certain embodiments provide therapeutic compositions comprising at least one compound disclosed herein in combination with one or more additional agents for the treatment of dopamine receptor-mediated disorders.
  • Isotopic hydrogen can be introduced into a compound as disclosed herein by synthetic techniques that employ deuterated reagents, whereby incorporation rates are pre-determined; and/or by exchange techniques, wherein incorporation rates are determined by equilibrium conditions, and may be highly variable depending on the reaction conditions.
  • Synthetic techniques where tritium or deuterium is directly and specifically inserted by tritiated or deuterated reagents of known isotopic content, may yield high tritium or deuterium abundance, but can be limited by the chemistry required.
  • Exchange techniques on the other hand, may yield lower tritium or deuterium incorporation, often with the isotope being distributed over many sites on the molecule.
  • the compounds as disclosed herein can be prepared by methods known to one of skill in the art and routine modifications thereof, and/or following procedures similar to those described in the Example section herein and routine modifications thereof, and/or procedures found in WO 2006123356; WO 2008084499; WO 2007010557; WO 9415918; EP 1568689; US 20050159605; WO 2005040115; Gallagher et al., /. Med. Chem. , 1985, 28(10), 1533-6, which are hereby incorporated in their entirety, and references cited therein and routine modifications thereof.
  • Compounds as disclosed herein can also be prepared as shown in any of the following schemes and routine modifications thereof. [00102]
  • the following schemes can be used to practice the present invention. Any position shown as hydrogen may optionally be replaced with deuterium.
  • Compound 1 is treated with an appropriate reducing agent, such as sodium borohydride, in the presence of an appropriate acid, such as methanesulfonic acid, in an appropriate solvent, such as tetrahydrofuran, to give compound 2.
  • an appropriate hydroxyl activating agent such as methanesulfonyl chloride
  • an appropriate base such as triethylamine
  • an appropriate solvent such as dichloromethane
  • Compound 4 is treated with an appropriate acid, such as sulfuric acid, in an appropriate solvent, such as a mixture of acetic acid and water, to give compound 5.
  • Compound 5 is treated with an appropriate reducing agent, such as sodium borohydride, in the presence of an appropriate acid, such as methanesulfonic acid, in an appropriate solvent, such as tetrahydrofuran, to give compound 6.
  • Compound 6 is reacted with an appropriate hydroxyl activating agent, such as para- toluenesulfonyl chloride, in the presence of an appropriate base, such as triethylamine, in an appropriate solvent, such as dichloromethane, to give compound 7.
  • Compound 7 is reacted with compound 8 in an appropriate solvent, such as water, to give compound 9.
  • Compound 9 is reacted with an appropriate dialkyl oxalate, such as diethyl oxalate, in the presence of an appropriate base, such as sodium ethoxide, in an appropriate solvent, such as a mixture of ethanol and tetrahydrofuran, to give compound 10.
  • an appropriate base such as sodium ethoxide
  • an appropriate solvent such as a mixture of ethanol and tetrahydrofuran
  • Compound 10 is reacted with hydrogen peroxide in the presence of an appropriate base, such as sodium hydroxide, in an appropriate solvent, such as water, to give compound 11.
  • Compound 11 is reacted with an appropriate reducing agent, such as hydrogen gas and an appropriate catalyst, such as palladium on carbon, in an appropriate solvent, such as methanol, to give a compound of formula I.
  • Deuterium can be incorporated to different positions synthetically, according to the synthetic procedures as shown in Scheme I, by using appropriate deuterated intermediates.
  • compound 1 with the corresponding deuterium substitutions can be used.
  • sodium borodeuteride can be used.
  • compound 8 with the corresponding deuterium substitutions can be used.
  • Deuterium can be incorporated to various positions having an exchangeable proton, such as the indolinone N-H, via proton-deuterium equilibrium exchange.
  • this proton may be replaced with deuterium selectively or non-selectively through a proton-deuterium exchange method known in the art.
  • Liver microsomal stability assays are conducted at 1 mg per mL liver microsome protein with an NADPH-generating system in 2% NaHC ⁇ 3 (2.2 mM NADPH, 25.6 mM glucose 6-phosphate, 6 units per mL glucose 6-phosphate dehydrogenase and 3.3 mM MgCl 2 ).
  • Test compounds are prepared as solutions in 20% acetonitrile-water and added to the assay mixture (final assay concentration 5 microgram per mL) and incubated at 37 0 C. Final concentration of acetonitrile in the assay should be ⁇ 1%.
  • cytochrome P 4 50 enzymes In vitro metabolism using human cytochrome P 4 50 enzymes [00110]
  • the cytochrome P 450 enzymes are expressed from the corresponding human cDNA using a baculovirus expression system (BD Biosciences, San Jose, CA).
  • reaction is stopped by the addition of an appropriate solvent (e.g., acetonitrile, 20% trichloroacetic acid, 94% acetonitrile/6% glacial acetic acid, 70% perchloric acid, 94% acetonitrile/6% glacial acetic acid) and centrifuged (10,000 g) for 3 min. The supernatant is analyzed by HPLC/MS/MS.
  • an appropriate solvent e.g., acetonitrile, 20% trichloroacetic acid, 94% acetonitrile/6% glacial acetic acid, 70% perchloric acid, 94% acetonitrile/6% glacial acetic acid
  • Monoamine Oxidase A Inhibition and Oxidative Turnover [00111] The procedure is carried out using the methods described by Weyler, Journal of Biological Chemistry 1985, 260, 13199-13207, which is hereby incorporated by reference in its entirety. Monoamine oxidase A activity is measured spectrophotometrically by monitoring the increase in absorbance at 314 nm on oxidation of kynuramine with formation of 4-hydroxyquinoline. The measurements are carried out, at 30 0 C, in 5OmM NaP 1 buffer, pH 7.2, containing 0.2% Triton X-100 (monoamine oxidase assay buffer), plus 1 mM kynuramine, and the desired amount of enzyme in 1 mL total volume.

Landscapes

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

Abstract

La présente invention concerne de nouveaux modulateurs à base d’indolinone du récepteur de la dopamine, leurs compositions pharmaceutiques et leurs procédés d’utilisation.
PCT/US2009/065637 2008-11-24 2009-11-24 Modulateurs à base d'indolinone du récepteur de la dopamine WO2010060070A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11725008P 2008-11-24 2008-11-24
US61/117,250 2008-11-24

Publications (2)

Publication Number Publication Date
WO2010060070A2 true WO2010060070A2 (fr) 2010-05-27
WO2010060070A3 WO2010060070A3 (fr) 2010-09-23

Family

ID=42196902

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/065637 WO2010060070A2 (fr) 2008-11-24 2009-11-24 Modulateurs à base d'indolinone du récepteur de la dopamine

Country Status (2)

Country Link
US (1) US20100130582A1 (fr)
WO (1) WO2010060070A2 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8652527B1 (en) 2013-03-13 2014-02-18 Upsher-Smith Laboratories, Inc Extended-release topiramate capsules
US9101545B2 (en) 2013-03-15 2015-08-11 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016109359A1 (fr) * 2014-12-29 2016-07-07 Auspex Pharmaceuticals, Inc. Modulateurs de récepteurs de la mélatonine à base de cyclopropyl dihydrobenzofurane

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4452808A (en) * 1982-12-07 1984-06-05 Smithkline Beckman Corporation 4-Aminoalkyl-2(3H)-indolones

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BLOOMER, J.C. ET AL.: 'In vitro identification of the P450 enzymes responsible for the metabolism of Ropinirole' DRUG METABOLISM AND DISPOSITION vol. 25, no. 7, 1997, pages 840 - 844 *
FOSTER, A. B.: 'Deuterium isotope effects in studies of drug metabolism' TRENDS IN PHARMACOLOGICAL SCIENCES vol. 5, 1984, pages 524 - 527 *
KUSHNER,D.J. ET AL.: 'Pharmacological uses and perspectives of heavy water and deuterated compounds' CANADIAN JOURNAL OF PHYSIOLOGY AND PHARMACOLOGY vol. 77, 1999, pages 79 - 88 *
REAVILL, C. ET AL.: 'Comparative Pharmacological Study of Ropinirole and its Metabolites in Rats' JOURNAL OF PHARMACY AND PHARMACOLOGY vol. 52, 2000, pages 1129 - 1135 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8652527B1 (en) 2013-03-13 2014-02-18 Upsher-Smith Laboratories, Inc Extended-release topiramate capsules
US8889190B2 (en) 2013-03-13 2014-11-18 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
US10363224B2 (en) 2013-03-13 2019-07-30 Upsher-Smith Laboratories, Llc Extended-release topiramate capsules
US9101545B2 (en) 2013-03-15 2015-08-11 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
US9555005B2 (en) 2013-03-15 2017-01-31 Upsher-Smith Laboratories, Inc. Extended-release topiramate capsules
US10172878B2 (en) 2013-03-15 2019-01-08 Upsher-Smith Laboratories, Llc Extended-release topiramate capsules

Also Published As

Publication number Publication date
WO2010060070A3 (fr) 2010-09-23
US20100130582A1 (en) 2010-05-27

Similar Documents

Publication Publication Date Title
US9260424B2 (en) 4,6-diaminopyrimidine stimulators of soluble guanylate cyclase
US20110206780A1 (en) Morphinan modulators of nmda receptors, sigma1 receptors, sigma2 receptors, and/or a3b4 nicotinic receptors
US20100291151A1 (en) 1-methylpyrazole modulators of substance p, calcitonin gene-related peptide, adrenergic receptor, and/or 5-ht receptor
US20110082151A1 (en) Sulfonylurea modulators of endothelin receptor
WO2011060363A2 (fr) Modulateurs des récepteurs d2 et/ou des récepteurs d3 à base de cyclohexylurée
US20100143507A1 (en) Carboxylic acid inhibitors of histone deacetylase, gaba transaminase and sodium channel
US20110257260A1 (en) 3,4-methylenedioxyphenyl inhibitors of gaba aminotransferase and/or gaba reuptake transporter inhibitor
US20100105755A1 (en) Substituted benzamide modulators of dopamine receptor
US20100113496A1 (en) Piperidine modulators of vmat2
WO2010057088A2 (fr) Modulateurs de pyrrolidinyle de récepteurs nicotiniques d'acétylcholine
US20100076074A1 (en) Carbamate reducers of skeletal muscle tension
US20100286124A1 (en) Prop-2-yn-1-amine inhibitors of monoamine oxidase type b
US20100075950A1 (en) Phenylpropanone modulators of dopamine receptor
US20110091459A1 (en) Imidazole modulators of muscarinic acetylcholine receptor m3
US20100124541A1 (en) Hydroxyadamantyl inhibitors of dipeptidylpeptidase iv
US20100130582A1 (en) Indolinone modulators of dopamine receptor
WO2010068717A2 (fr) Désactivateurs pyrazolinone de radicaux libres
US20100113405A1 (en) Methylindazole modulators of 5-ht3 receptors
US20100317655A1 (en) Sulfonamide inhibitors of carbonic anhydrase
WO2010060041A2 (fr) Inhibiteurs des canaux potassiques sensibles à l'atp à base de phénylalanine-amide
WO2010056741A2 (fr) Inhibiteurs de cyclooxygénase à base d'acide phénylacétique
US20100130617A1 (en) Ethanolamine modulators of nmda receptor and muscarinic acetylcholine receptor
US20100113478A1 (en) Indolone modulators of 5-ht3 receptor
US20100093758A1 (en) Pyridine sulfonamide modulators of endothelin-a receptor
US20100137332A1 (en) Piperazine modulators of nk-1 receptors

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: 09828369

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09828369

Country of ref document: EP

Kind code of ref document: A2