US20150202198A1 - Novel Cabergoline Derivatives - Google Patents

Novel Cabergoline Derivatives Download PDF

Info

Publication number
US20150202198A1
US20150202198A1 US14/670,953 US201514670953A US2015202198A1 US 20150202198 A1 US20150202198 A1 US 20150202198A1 US 201514670953 A US201514670953 A US 201514670953A US 2015202198 A1 US2015202198 A1 US 2015202198A1
Authority
US
United States
Prior art keywords
alkyl
substituted
hydrogen
compounds
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/670,953
Inventor
Jian Zhang
Thomas A. Armer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MAP Pharmaceuticals Inc
Original Assignee
MAP 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 MAP Pharmaceuticals Inc filed Critical MAP Pharmaceuticals Inc
Priority to US14/670,953 priority Critical patent/US20150202198A1/en
Publication of US20150202198A1 publication Critical patent/US20150202198A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/48Ergoline derivatives, e.g. lysergic acid, ergotamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D457/00Heterocyclic compounds containing indolo [4, 3-f, g] quinoline ring systems, e.g. derivatives of ergoline, of the formula:, e.g. lysergic acid
    • C07D457/04Heterocyclic compounds containing indolo [4, 3-f, g] quinoline ring systems, e.g. derivatives of ergoline, of the formula:, e.g. lysergic acid with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 8
    • C07D457/06Lysergic acid amides

Definitions

  • novel cabergoline analogs and compositions thereof are provided herein.
  • methods of treatment, prevention, or amelioration of a variety of medical disorders such as, for example, migraine and Parkinson's disease using the compounds and compositions disclosed herein.
  • methods of agonizing dopaminergic D 2 receptors and/or antagonizing or inhibiting activity of receptors such as the 5-HT 2 serotonin receptors using the compounds and compositions disclosed herein.
  • methods of agonizing receptors such as, for example, the 5-HT 1D and/or the 5-HT 1B receptor, without agonizing (or with very weak agonism of) the 5-HT 2B receptor using the compounds and compositions disclosed herein.
  • Cabergoline depicted below was approved for the treatment of Parkinson's disease but was subsequently withdrawn from the U.S. market because of undesirable agonization of the 5-HT 2B receptor which leads to cardiac and non-cardiac fibrosis.
  • an ideal anti-Parkinson's agent will be a selective agonist for dopaminergic D 2 receptor.
  • Weak to modest agonist activity on the 5-HT1B and/or 5-HT1D receptors may also be desirable.
  • 5-HT2 receptor antagonism may also be desirable for the treatment of Parkinson's disease symptoms and/or for the reduction of undesirable side-effects, such as cardiac or non-cardiac fibrosis and psychiatric side-effects (Newman-Trancredi, J Pharmacology and Experimental Therapies (2002) 303(2):815-822).
  • anti-Parkinson's agents may have a relatively short half-life (i.e., 1-5 hours) or a longer half-life (i.e., 20 hours or longer).
  • cabergoline analogs which retain activity against central nervous disorders, such as, for example, Parkinson's disease and lack agonist activity, or very weak agonist activity, against the 5-HT 2 receptors, including 5-HT 2B and the 5-HT 2C receptors.
  • cabergoline derivatives which address these and other needs.
  • cabergoline derivatives described herein include compounds of Formula (I) or (II):
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms;
  • R 2 is alkyl, substituted alkyl, acyl, substituted acyl, halo, heteroalkyl, substituted heteroalkyl, —NO 2 , —N 3 , —OH, —S(O) k R 100 , —OR 101 , —NR 102 R 103 , —CONR 104 R 105 , —CO 2 R 106 or —O 2 CR 107 ;
  • R 5 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms, arylalkyl or substituted arylalkyl;
  • R 7 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms, arylalkyl or substituted arylalkyl;
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms, heteroalkyl, substituted heteroalkyl, arylalkyl, substituted arylalkyl, heteroaryl, substituted heteroaryl or R 4 and R 5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring
  • R 11 is hydrogen, (C 1 -C 3 ) alkyl or (C 1 -C 3 ) alkyl substituted with one or more fluorine atoms;
  • R 100 -R 107 are independently hydrogen, alkyl, substituted alkyl, acyl, substituted acyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl or substituted heteroarylalkyl;
  • k 0, 1 or 2;
  • n 0, 1, 2 or 3.
  • compositions which include the compounds provided herein and a vehicle.
  • Methods of treating, preventing, or ameliorating symptoms of medical disorders such as, for example, central nervous system disorders, migraine and Parkinson's disease are also provided herein.
  • therapeutically effective amounts of the compounds or compositions thereof are administered to a subject.
  • antagonizing receptors such as, for example, the 5-HT 2A and 5-HT 2B with the compounds and compositions described herein are also provided herein.
  • Methods of agonizing dopaminergic D 2 receptors using the compounds and compositions disclosed herein are also provided.
  • therapeutically effective amounts of the compounds or compositions are administered.
  • methods for selectively agonizing dopaminergic D 2 receptors are provided.
  • methods of antagonizing or inhibiting (or not agonizing) activity of the 5-HT 2 receptors are also provided.
  • methods and compounds for both selectively agonizing dopaminergic D 2 receptors and antagonizing (or not agonizing/very weak agonist activity) 5-HT 2 receptors at the same time are provided. Such methods and compounds may be desirable as agents for treating one or more symptoms of Parkinson's disease.
  • Alkyl by itself or as part of another substituent, refers to a saturated or unsaturated, branched, straight-chain or cyclic monovalent hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane, alkene or alkyne.
  • Typical alkyl groups include, but are not limited to, methyl; ethyls such as ethanyl, ethenyl, ethynyl; propyls such as propan-1-yl, propan-2-yl, cyclopropan-1-yl, prop-1-en-1-yl, prop-1-en-2-yl, prop-2-en-1-yl (allyl), cycloprop-1-en-1-yl; cycloprop-2-en-1-yl, prop-1-yn-1-yl, prop-2-yn-1-yl, etc.; butyls such as butan-1-yl, butan-2-yl, 2-methyl-propan-1-yl, 2-methyl-propan-2-yl, cyclobutan-1-yl, but-1-en-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-2-yl, buta-1,
  • alkyl is specifically intended to include groups having any degree or level of saturation, i.e., groups having exclusively single carbon-carbon bonds, groups having one or more double carbon-carbon bonds, groups having one or more triple carbon-carbon bonds and groups having mixtures of single, double and triple carbon-carbon bonds. Where a specific level of saturation is intended, the expressions “alkanyl,” “alkenyl,” and “alkynyl” are used.
  • an alkyl group comprises from 1 to 20 carbon atoms (C 1 -C 20 alkyl). In other embodiments, an alkyl group comprises from 1 to 10 carbon atoms (C 1 -C 10 alkyl). In still other embodiments, an alkyl group comprises from 1 to 6 carbon atoms (C 1 -C 6 alkyl).
  • Alkanyl by itself or as part of another substituent, refers to a saturated branched, straight-chain or cyclic alkyl radical derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane.
  • Typical alkanyl groups include, but are not limited to, methanyl; ethanyl; propanyls such as propan-1-yl, propan-2-yl (isopropyl), cyclopropan-1-yl, etc.; butanyls such as butan-1-yl, butan-2-yl (sec-butyl), 2-methyl-propan-1-yl (isobutyl), 2-methyl-propan-2-yl (t-butyl), cyclobutan-1-yl, etc.; and the like.
  • Alkenyl by itself or as part of another substituent, refers to an unsaturated branched, straight-chain or cyclic alkyl radical having at least one carbon-carbon double bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkene.
  • the group may be in either the cis or trans conformation about the double bond(s).
  • Typical alkenyl groups include, but are not limited to, ethenyl; propenyls such as prop-1-en-1-yl, prop-1-en-2-yl, prop-2-en-1-yl (allyl), prop-2-en-2-yl, cycloprop-1-en-1-yl; cycloprop-2-en-1-yl; butenyls such as but-1-en-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-1-yl, but-2-en-1-yl, but-2-en-2-yl, buta-1,3-dien-1-yl, buta-1,3-dien-2-yl, cyclobut-1-en-1-yl, cyclobut-1-en-3-yl, cyclobuta-1,3-dien-1-yl, etc.; and the like.
  • Alkynyl by itself or as part of another substituent refers to an unsaturated branched, straight-chain or cyclic alkyl radical having at least one carbon-carbon triple bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkyne.
  • Typical alkynyl groups include, but are not limited to, ethynyl; propynyls such as prop-1-yn-1-yl, prop-2-yn-1-yl, etc.; butynyls such as but-1-yn-1-yl, but-1-yn-3-yl, but-3-yn-1-yl, etc.; and the like.
  • “Acyl” by itself or as part of another substituent refers to a radical —C(O)R 400 , where R 400 is hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroalkyl, substituted heteroalkyl, heteroarylalkyl or substituted heteroarylalkyl as defined herein.
  • Representative examples include, but are not limited to formyl, acetyl, cyclohexylcarbonyl, cyclohexylmethylcarbonyl, benzoyl, benzylcarbonyl and the like.
  • Aryl by itself or as part of another substituent, refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system, as defined herein.
  • Typical aryl groups include, but are not limited to, groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phen
  • an aryl group comprises from 6 to 20 carbon atoms (C 6 -C 20 aryl). In other embodiments, an aryl group comprises from 6 to 15 carbon atoms (C 6 -C 15 aryl). In still other embodiments, an aryl group comprises from 6 to 15 carbon atoms (C 6 -C 10 aryl).
  • Arylalkyl by itself or as part of another substituent, refers to an acyclic alkyl group in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp 3 carbon atom, is replaced with an aryl group as, as defined herein.
  • Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, 2-phenylethen-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, 2-naphthylethen-1-yl, naphthobenzyl, 2-naphthophenylethan-1-yl and the like.
  • an arylalkyl group is (C 6 -C 30 ) arylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is (C 1 -C 10 ) alkyl and the aryl moiety is (C 6 -C 20 ) aryl.
  • an arylalkyl group is (C 6 -C 20 ) arylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is (C 1 -C 8 ) alkyl and the aryl moiety is (C 6 -C 12 ) aryl.
  • an arylalkyl group is (C 6 -C 15 ) arylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is (C 1 -C 5 ) alkyl and the aryl moiety is (C 6 -C 10 ) aryl.
  • Compounds refers to compounds encompassed by structural formulae disclosed herein and includes any specific compounds within these formulae whose structure is disclosed herein. Compounds may be identified either by their chemical structure and/or chemical name. When the chemical structure and chemical name conflict, the chemical structure is determinative of the identity of the compound.
  • the compounds described herein may contain one or more chiral centers and/or double bonds and therefore, may exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), enantiomers or diastereomers.
  • the chemical structures depicted herein encompass all possible enantiomers and stereoisomers of the illustrated compounds including the stereoisomerically pure form (e.g., geometrically pure, enantiomerically pure or diastereomerically pure) and enantiomeric and stereoisomeric mixtures.
  • Enantiomeric and stereoisomeric mixtures can be resolved into their component enantiomers or stereoisomers using separation techniques or chiral synthesis techniques well known to the skilled artisan.
  • the compounds may also exist in several tautomeric forms including the enol form, the keto form and mixtures thereof. Accordingly, the chemical structures depicted herein encompass all possible tautomeric forms of the illustrated compounds.
  • the compounds described also include isotopically labeled compounds where one or more atoms have an atomic mass different from the atomic mass conventionally found in nature.
  • isotopes that may be incorporated into the compounds described herein include, but are not limited to, 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 35 S, etc. In general, it should be understood that all isotopes of any of the elements comprising the compounds described herein may be found in these compounds.
  • Compounds may exist in unsolvated or unhydrated forms as well as solvated forms, including hydrated forms and as N-oxides. In general, compounds may be hydrated, solvated or N-oxides. Certain compounds may exist in multiple crystalline or amorphous forms.
  • Heteroalkyl “Heteroalkanyl,” “Heteroalkenyl” and “Heteroalkynyl,” by themselves or as part of other substituents, refer to alkyl, alkanyl, alkenyl and alkynyl groups, respectively, in which one or more of the carbon atoms (and optionally any associated hydrogen atoms), are each, independently of one another, replaced with the same or different heteroatoms or heteroatomic groups.
  • Typical heteroatoms or heteroatomic groups which can replace the carbon atoms include, but are not limited to, —O—, —S—, —N—, —Si—, —NH—, —S(O)—, —S(O) 2 —, —S(O)NH—, —S(O) 2 NH— and the like and combinations thereof.
  • the heteroatoms or heteroatomic groups may be placed at any interior position of the alkyl, alkenyl or alkynyl groups.
  • Typical heteroatomic groups which can be included in these groups include, but are not limited to, —O—, —S—, —O—O—, —S—S—, —O—S—, —NR 501 R 502 —, ⁇ N—N ⁇ , —N ⁇ N—, —N ⁇ N—NR 503 R 404 , —PR 505 —, —P(O) 2 —, —POR 506 —, —O—P(O) 2 —, —SO—, —SO 2 —, —SnR 507 R 508 — and the like, where R 501 , R 502 , R 503 , R 504 , R 505 , R 506 , R 507 and R 508 — are independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, cycloalkyl, substituted cycloalkyl,
  • Heteroaryl by itself or as part of another substituent, refers to a monovalent heteroaromatic radical derived by the removal of one hydrogen atom from a single atom of a parent heteroaromatic ring systems, as defined herein.
  • Typical heteroaryl groups include, but are not limited to, groups derived from acridine, ⁇ -carboline, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole,
  • the heteroaryl group comprises from 5 to 20 ring atoms (5-20 membered heteroaryl). In other embodiments, the heteroaryl group comprises from 5 to 10 ring atoms (5-10 membered heteroaryl).
  • Exemplary heteroaryl groups include those derived from furan, thiophene, pyrrole, benzothiophene, benzofuran, benzimidazole, indole, pyridine, pyrazole, quinoline, imidazole, oxazole, isoxazole and pyrazine.
  • Heteroarylalkyl by itself or as part of another substituent refers to an acyclic alkyl group in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp 3 carbon atom, is replaced with a heteroaryl group. Where specific alkyl moieties are intended, the nomenclature heteroarylalkanyl, heteroarylakenyl and/or heteroarylalkynyl is used.
  • the heteroarylalkyl group is a 6-21 membered heteroarylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the heteroarylalkyl is (C 1 -C 6 ) alkyl and the heteroaryl moiety is a 5-15-membered heteroaryl.
  • the heteroarylalkyl is a 6-13 membered heteroarylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety is (C 1 -C 3 ) alkyl and the heteroaryl moiety is a 5-10 membered heteroaryl.
  • “Hydrates” refers to incorporation of water into to the crystal lattice of a compound described herein, in stochiometric proportions, resulting in the formation of an adduct.
  • Methods of making hydrates include, but are not limited to, storage in an atmosphere containing water vapor, dosage forms that include water, or routine pharmaceutical processing steps such as, for example, crystallization (i.e., from water or mixed aqueous solvents), lyophilization, wet granulation, aqueous film coating, or spray drying. Hydrates may also be formed, under certain circumstances, from crystalline solvates upon exposure to water vapor, or upon suspension of the anhydrous material in water.
  • Hydrates may also crystallize in more than one form resulting in hydrate polymorphism. See e.g., (Guillory, K., Chapter 5, pp. 202-205 in Polymorphism in Pharmaceutical Solids , (Brittain, H. ed.), Marcel Dekker, Inc., New York, N.Y., 1999).
  • the above methods for preparing hydrates are well within the ambit of those of skill in the art, are completely conventional and do not require any experimentation beyond what is typical in the art.
  • Hydrates may be characterized and/or analyzed by methods well known to those of skill in the art such as, for example, single crystal X-Ray diffraction, X-Ray powder diffraction, Polarizing optical microscopy, thermal microscopy, thermogravimetry, differential thermal analysis, differential scanning calorimetry, IR spectroscopy, Raman spectroscopy and NMR spectroscopy. (Brittain, H., Chapter 6, pp. 205-208 in Polymorphism in Pharmaceutical Solids , (Brittain, H. ed.), Marcel Dekker, Inc. New York, 1999).
  • Parent aromatic Ring System refers to an unsaturated cyclic or polycyclic ring system having a conjugated ⁇ electron system. Specifically included within the definition of “parent aromatic ring system” are fused ring systems in which one or more of the rings are aromatic and one or more of the rings are saturated or unsaturated, such as, for example, fluorene, indane, indene, phenalene, etc.
  • Typical parent aromatic ring systems include, but are not limited to, accanthrylene, accnaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene and the like.
  • Preventing refers to a reduction in risk of acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a patient that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease).
  • preventing refers to reducing symptoms of the disease by taking the compound in a preventative fashion.
  • the application of a therapeutic for preventing or prevention of a disease of disorder is known as ‘prophylaxis.’
  • the compounds provided herein provide superior prophylaxis because of lower long term side effects over long time periods.
  • Prodrug refers to a derivative of a drug molecule that requires a transformation within the body to release the active drug. Prodrugs are frequently (though not necessarily) pharmacologically inactive until converted to the parent drug.
  • “Promoiety” refers to a form of protecting group that when used to mask a functional group within a drug molecule converts the drug into a prodrug. Typically, the promoiety will be attached to the drug via bond(s) that are cleaved by enzymatic or non-enzymatic means in vivo.
  • Salt refers to a salt of a compound, which possesses the desired pharmacological activity of the parent compound.
  • Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesul
  • Solidvates refers to incorporation of solvents into to the crystal lattice of a compound described herein, in stochiometric proportions, resulting in the formation of an adduct.
  • Methods of making solvates include, but are not limited to, storage in an atmosphere containing a solvent, dosage forms that include the solvent, or routine pharmaceutical processing steps such as, for example, crystallization (i.e., from solvent or mixed solvents) vapor diffusion, etc.
  • Solvates may also be formed, under certain circumstances, from other crystalline solvates or hydrates upon exposure to the solvent or upon suspension material in solvent. Solvates may crystallize in more than one form resulting in solvate polymorphism.
  • Solvates may be characterized and/or analyzed by methods well known to those of skill in the art such as, for example, single crystal X-Ray diffraction, X-Ray powder diffraction, Polarizing optical microscopy, thermal microscopy, thermogravimetry, differential thermal analysis, differential scanning calorimetry, IR spectroscopy, Raman spectroscopy and NMR spectroscopy. (Brittain, H., Chapter 6, pp. 205-208 in Polymorphism in Pharmaceutical Solids , (Brittain, H. ed.), Marcel Dekker, Inc. New York, 1999).
  • “Substituted,” when used to modify a specified group or radical, means that one or more hydrogen atoms of the specified group or radical are each, independently of one another, replaced with the same or different substituent(s).
  • Substituent groups useful for substituting saturated carbon atoms in the specified group or radical include, but are not limited to —R a , halo, —O ⁇ , ⁇ OR b , —SR b , —S ⁇ , ⁇ S, —NR c R c , ⁇ NR b , ⁇ N—OR b , trihalomethyl, —CF 3 , —CN, —OCN, —SCN, —NO, —NO 2 , ⁇ N 2 , —N 3 , —S(O) 2 R b , —S(O) 2 NR b , —S(O) 2 O ⁇ , —S(O) 2 OR b , —OS(O) 2
  • substituent groups useful for substituting unsaturated carbon atoms in the specified group or radical include, but are not limited to, —R a , halo, —O ⁇ , —OR b , —SR b , —S ⁇ , —NR c R c , trihalomethyl, —CF 3 , —CN, —OCN, —SCN, —NO, —NO 2 , —N 3 , —S(O) 2 R b , —S(O) 2 O ⁇ , —S(O) 2 OR b , —OS(O) 2 R b , —OS(O) 2 O ⁇ , —OS(O) 2 OR b , —P(O)(O ⁇ ) 2 , —P(O)(OR b )(O ⁇ ), —P(O)(OR b )(OR b ), —C(O)R b ,
  • Substituent groups useful for substituting nitrogen atoms in heteroalkyl and cycloheteroalkyl groups include, but are not limited to, —R a , —O ⁇ , —OR b , —SR b , —S ⁇ , —NR c R c , trihalomethyl, —CF 3 , —CN, —NO, —NO 2 , —S(O) 2 R b , —S(O) 2 O ⁇ , —S(O) 2 OR b , —OS(O) 2 R b , —OS(O) 2 O ⁇ , —OS(O) 2 OR b , —P(O)(O ⁇ ) 2 , —P(O)(OR b )(O ⁇ ), —P(O)(OR b )(OR b ), —C(O)R b , —C(S)R b ,
  • substituent groups from the above lists useful for substituting other specified groups or atoms will be apparent to those of skill in the art.
  • the substituents used to substitute a specified group can be further substituted, typically with one or more of the same or different groups selected from the various groups specified above. In some embodiments, substituents are limited to the groups above.
  • Subject refers to a vertebrate, preferably a mammal Mammals include, but are not limited to, murines, rodents, simians, humans, farm animals, sport animals and pets.
  • Treating” or “treatment” of any disease or disorder refers, in some embodiments, to ameliorating the disease or disorder (i.e., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof,). Treatment can also refer to the lessening the severity and/or the duration of one or more symptoms of a disease or disorder. Treatment may also be considered to include preemptive or prophylactic administration to ameliorate, arrest or prevent the development of the disease or at least one of the clinical symptoms. Treatment can also refer to the lessening of the severity and/or the duration of one or more symptoms of a disease or disorder. In a further feature, the treatment rendered has lower potential for long term side effects over multiple years.
  • treating or “treatment” refers to ameliorating at least one physical parameter, which may not be discernible by the patient.
  • “treating” or “treatment” refers to inhibiting the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter) or both.
  • “treating” or “treatment” refers to delaying the onset of the disease or disorder.
  • “Therapeutically effective amount” means the amount of a compound that, when administered to a patient for treating a disease, is sufficient to effect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, adsorption, distribution, metabolism and excretion etc., of the patient to be treated.
  • Vehicle refers to a diluent, excipient or carrier with which a compound is administered to a subject.
  • the vehicle is pharmaceutically acceptable.
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms;
  • R 2 is alkyl, substituted alkyl, acyl, substituted acyl, halo, heteroalkyl, substituted heteroalkyl, —NO 2 , —N 3 , —OH, —S(O) k R 100 , —OR 101 , —NR 102 R 103 , —CONR 104 R 105 , —CO 2 R 106 or —O 2 CR 107 ;
  • R 5 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms, arylalkyl or substituted arylalkyl;
  • R 7 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms, arylalkyl or substituted arylalkyl;
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms, heteroalkyl, substituted heteroalkyl, arylalkyl, substituted arylalkyl, heteroaryl, substituted heteroaryl or R 4 and R 5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring
  • R 11 is hydrogen, (C 1 -C 3 ) alkyl or (C 1 -C 3 ) alkyl substituted with one or more fluorine atoms;
  • R 100 -R 107 are independently hydrogen, alkyl, substituted alkyl, acyl, substituted acyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl or substituted heteroarylalkyl;
  • k 0, 1 or 2;
  • n 0, 1, 2 or 3.
  • R 1 and R 11 are not both hydrogen.
  • R 11 when R 11 is hydrogen, R 1 is substituted (C 1 -C 4 ) alkyl with one or more fluorine atoms.
  • R 11 when R 11 is hydrogen, R 1 is (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms.
  • R 11 when R 1 is (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms.
  • R 11 is (C 1 -C 3 ) alkyl substituted with one or more fluorine atoms.
  • R 11 is —CF 3 .
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms. In other embodiments, R 1 is hydrogen, —CH 3 or —CF 3 .
  • R 2 is alkyl, acyl, halo, —NO 2 , —OH, —S(O) k R 100 , —OR 101 , —NR 102 R 103 , —CONR 104 R 105 , —CO 2 R 106 or —O 2 CR 107 .
  • R 2 is alkyl, acyl, halo, —NO 2 , —OH, —S(O) k R 100 , —OR 101 , —NR 102 R 103 , —CONR 104 R 105 , —CO 2 R 106 or —O 2 CR 107 and n is 1.
  • R 2 is alkyl, halo or —OR 101 and n is 1. In still other embodiments, n is 0.
  • R 5 is hydrogen or (C 1 -C 3 ) alkyl. In other embodiments, R 5 is hydrogen, methyl or allyl. In still other embodiments, R 5 is methyl. In still other embodiments, R 5 is allyl.
  • R 7 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms. In other embodiments, R 7 is hydrogen or (C 1 -C 4 ) alkyl substituted with one dialkyl amino group.
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms, or R 4 and R 5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring.
  • R 8 and R 9 are independently hydrogen or (C 1 -C 4 ) alkyl. In other embodiments, R 8 is hydrogen and R 9 is (C 1 -C 4 ) alkyl.
  • R 11 is (C 1 -C 3 ) alkyl substituted with one or more fluorine atoms. In other embodiments, R 11 is —CF 3 .
  • R 100 -R 122 are independently hydrogen, alkyl, or substituted alkyl, acyl or substituted acyl. In some embodiments, R 100 -R 122 are independently hydrogen or alkyl.
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • R 2 is alkyl, acyl, halo, —NO 2 , —OH, —S(O) k R 100 , —OR 101 , —NR 102 R 103 , —CONR 104 R 105 , —CO 2 R 106 or —O 2 CR 107 and n is 1
  • R 5 is hydrogen or (C 1 -C 3 ) alkyl
  • R 7 is hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) substituted alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl, (C 1 -C 4
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • n is 0,
  • R 5 is hydrogen or (C 1 -C 3 ) alkyl
  • R 7 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms, or R 4 and R 5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring and
  • R 11 is (C 1 -C 3 ) alkyl substituted with
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • R 2 is alkyl, acyl, halo, —NO 2 , —OH, —S(O) k R 100 , —OR 100 , —NR 102 R 103 , —CONR 104 R 105 , —CO 2 R 106 or —O 2 CR 107 and n is 1, fluoro, hydroxy or methoxy
  • R 5 is hydrogen or (C 1 -C 3 ) alkyl
  • R 7 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl, (
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • n is 0,
  • R 5 is hydrogen or (C 1 -C 3 ) alkyl
  • R 7 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl, (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms, or R 4 and R 5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring and
  • R 11 is (C 1 -C 3 ) alkyl substituted with
  • R 1 is hydrogen, —CH 3 or —CF 3
  • R 2 is alkyl, halo or —OR 101 and n is 1
  • R 5 is hydrogen, methyl or allyl
  • R 7 is hydrogen or (C 1 -C 4 ) alkyl substituted with one dimethyl amino group
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) substituted alkyl and R 11 is —CF 3 .
  • R 1 is hydrogen, —CH 3 or —CF 3
  • n is 0,
  • R 5 is hydrogen, methyl or allyl
  • R 7 is hydrogen or (C 1 -C 4 ) alkyl substituted with one dimethyl amino group
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) substituted alkyl and R 11 is —CF 3 .
  • R 1 is hydrogen, —CH 3 or —CF 3
  • R 2 is alkyl, halo or —OR 101 and n is 1
  • R 5 is hydrogen, methyl or allyl
  • R 7 is hydrogen or (C 1 -C 4 ) alkyl substituted with one dimethyl amino group
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) substituted alkyl and R 11 is —CF 3 .
  • R 1 is hydrogen, —CH 3 or —CF 3
  • n is 0,
  • R 5 is hydrogen, methyl or allyl
  • R 7 is hydrogen or (C 1 -C 4 ) alkyl substituted with one dimethyl amino group
  • R 8 and R 9 are independently hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) substituted alkyl and R 11 is —CF 3 .
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms. In other embodiments, R 1 is hydrogen, —CH 3 or —CF 3 . In still other embodiments, R 1 is —CH 3 or —CF 3 ,
  • R 5 is hydrogen or (C 1 -C 3 ) alkyl. In other embodiments, R 5 is hydrogen, methyl or allyl. In still other embodiments, R 5 is methyl or allyl.
  • R 2 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms.
  • R 7 is substituted (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one dialkyl amino group. In till other embodiments, R 7 is 3-dimethylaminopropyl.
  • R 9 is hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms. In other embodiments, R 9 is (C1-C4) alkyl or (C 1 -C 4 ) substituted alkyl. In still other embodiments, R 9 is (C 1 -C 4 ) alkyl. In still other embodiments, R 9 is ethyl.
  • R 11 is hydrogen or (C 1 -C 3 ) alkyl substituted with one or more fluorine atoms. In other embodiments, R 11 is —CF 3 .
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • R 5 is hydrogen or (C 1 -C 3 ) alkyl
  • R 7 is hydrogen, (C 1 -C 4 ) alkyl, (C 1 -C 4 ) substituted alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • R 9 is hydrogen, (C 1 -C 4 ) alkyl, substituted (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • R 11 is hydrogen or (C 1 -C 3 ) alkyl substituted with one or more fluorine atoms.
  • R 1 is hydrogen, —CH 3 or —CF 3
  • R 5 is hydrogen, methyl or allyl
  • R 7 is substituted (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one dialkyl amino group
  • R 9 is (C 1 -C 4 ) alkyl and R 11 is —CF 3
  • R 1 is —CH 3 or —CF 3
  • R 5 is methyl or allyl
  • R 9 is (C 1 -C 4 ) alkyl and R 11 is —CF 3 .
  • R 1 is hydrogen, (C 1 -C 4 ) alkyl or (C 1 -C 4 ) alkyl substituted with one or more fluorine atoms
  • R 5 is hydrogen or (C 1 -C 3 ) alkyl
  • R 11 is (C 1 -C 3 ) alkyl substituted with one or more fluorine atoms.
  • R 1 is hydrogen, —CH 3 or —CF 3
  • R 5 is hydrogen, methyl or allyl
  • R 11 is hydrogen or —CF 3
  • R 1 is hydrogen, R 5 is allyl
  • R 11 is —CF 3
  • R 1 is —CH 3 or —CF 3
  • R 5 is methyl or allyl
  • R 11 is —CF 3
  • R 1 is —CH 3 or —CF 3
  • R 5 is methyl or allyl
  • R 11 is —CF 3
  • R 1 is —CH 3 or —CF 3
  • R 5 is methyl or allyl
  • R 11 is —CF 3 .
  • Exemplary compounds of Formula (I) include the compounds depicted below.
  • Exemplary compounds of Formula (II) include, for example, the 8,9-dihydro analogues of the above depicted compounds of Formula (I).
  • R 1 is hydrogen, —CH 3 or —CF 3
  • R 11 is hydrogen or —CF 3
  • R 1 is —CH 3 or —CF 3 and R 11 is —CF 3 .
  • direct functionalization of 2-unsubstituted analogs of compounds of Formula (I) and (II) e.g., compounds of Formula (VII) and (VIII)
  • an alkyl halide under basic conditions can be used to provide the compounds of Formula (I) and (II).
  • acids (IX) and (X) which can be prepared by methods well known to those of skill in the art can be used provide compounds of Formulas (I) and (II).
  • compositions provided herein contain therapeutically effective amounts of one or more of the compounds provided herein that are useful in the prevention, treatment, or amelioration of one or more of the symptoms of diseases or disorders described herein and a vehicle.
  • Vehicles suitable for administration of the compounds provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration.
  • the compounds may be formulated as the sole active ingredient in the composition or may be combined with other active ingredients.
  • compositions contain one or more compounds provided herein.
  • the compounds are, in some embodiments, formulated into suitable preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as topical administration, transdermal administration and oral inhalation via nebulizers, pressurized metered dose inhalers and dry powder inhalers.
  • the compounds described above are formulated into compositions using techniques and procedures well known in the art (see, e.g., Ansel Introduction to Pharmaceutical Dosage Forms, Seventh Edition (1999).
  • compositions effective concentrations of one or more compounds or derivatives thereof is (are) mixed with a suitable vehicle.
  • the compounds may be derivatized as the corresponding salts, esters, enol ethers or esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, ion-pairs, hydrates or prodrugs prior to formulation, as described above.
  • concentrations of the compounds in the compositions are effective for delivery of an amount, upon administration that treats, leads to prevention, or amelioration of one or more of the symptoms of diseases or disorders described herein.
  • the compositions are formulated for single dosage administration. To formulate a composition, the weight fraction of a compound is dissolved, suspended, dispersed or otherwise mixed in a selected vehicle at an effective concentration such that the treated condition is relieved, prevented, or one or more symptoms are ameliorated.
  • the active compound is included in the vehicle in an amount sufficient to exert a therapeutically useful effect in the absence of undesirable side effects on the patient treated.
  • the therapeutically effective concentration may be predicted empirically by testing the compounds in in vitro and in vivo systems well known to those of skill in the art and then extrapolated therefrom for dosages for humans. Human doses are then typically fine-tuned in clinical trials and titrated to response.
  • the concentration of active compound in the composition will depend on absorption, inactivation and excretion rates of the active compound, the physicochemical characteristics of the compound, the dosage schedule, and amount administered as well as other factors known to those of skill in the art. For example, the amount that is delivered is sufficient to ameliorate one or more of the symptoms of diseases or disorders as described herein.
  • a therapeutically effective dosage should produce a serum concentration of active ingredient of from about 0.001 ng/ml to about 50-200 ⁇ g/ml.
  • the compositions in other embodiments, should provide a dosage of from about 0.0001 mg to about 70 mg of compound per kilogram of body weight per day.
  • Dosage unit forms are prepared to provide from about 0.01 mg, 0.1 mg or 1 mg to about 500 mg, 1000 mg or 5000 mg, and in some embodiments from about 10 mg to about 500 mg of the active ingredient or a combination of essential ingredients per dosage unit form.
  • the active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment is a function of the disease being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data or subsequent clinical testing. It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.
  • solubilizing compounds such as use of liposomes, prodrugs, complexation/chelation, nanoparticles, or emulsions or tertiary templating.
  • co-solvents such as dimethylsulfoxide (DMSO)
  • surfactants or surface modifiers such as TWEEN®
  • complexing agents such as cyclodextrin or dissolution by enhanced ionization (i.e. dissolving in aqueous sodium bicarbonate).
  • Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective compositions.
  • the resulting mixture may be a solution, suspension, emulsion or the like.
  • the form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected vehicle.
  • the effective concentration is sufficient for ameliorating the symptoms of the disease, disorder or condition treated and may be empirically determined
  • compositions are provided for administration to humans and animals in indication appropriate dosage forms, such as dry powder inhalers (DPIs), pressurized metered dose inhalers (pMDIs), nebulizers, tablets, capsules, pills, sublingual tapes/bioerodible strips, tablets or capsules, powders, granules, lozenges, lotions, salves, suppositories, fast melts, transdermal patches or other transdermal application devices/preparations, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the compounds or derivatives thereof.
  • DPIs dry powder inhalers
  • pMDIs pressurized metered dose inhalers
  • nebulizers tablets, capsules, pills, sublingual tapes/bioerodible strips, tablets or capsules
  • powders granules
  • lozenges powders, granules, lozenges, lotions, salves, sup
  • the therapeutically active compounds and derivatives thereof are, in some embodiments, formulated and administered in unit-dosage forms or multiple-dosage forms.
  • Unit-dose forms as used herein refer to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required vehicle. Examples of unit-dose forms include ampoules and syringes and individually packaged tablets or capsules. Unit-dose forms may be administered in fractions or multiples thereof.
  • a multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pints or gallons. Hence, multiple dose form is a multiple of unit-doses which are not segregated in packaging.
  • Liquid compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional adjuvants in a vehicle, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension, colloidal dispersion, emulsion or liposomal formulation.
  • a vehicle such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension, colloidal dispersion, emulsion or liposomal formulation.
  • composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • compositions containing active ingredient in the range of 0.005% to 100% with the balance made up from vehicle or carrier may be prepared. Methods for preparation of these compositions are known to those skilled in the art.
  • the contemplated compositions may contain 0.001%-100% active ingredient, in one embodiment 0.1-95%, in another embodiment 0.4-10%.
  • the compositions are lactose-free compositions containing excipients that are well known in the art and are listed, for example, in the U.S. Pharmacopeia (USP) 25-NF20 (2002).
  • lactose-free compositions contain active ingredients, a binder/filler, and a lubricant in compatible amounts.
  • Particular lactose-free dosage forms contain active ingredients, microcrystalline cellulose, pre-gelatinized starch, and magnesium stearate.
  • anhydrous compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds.
  • water e.g., 5%
  • water e.g., 5%
  • water and heat accelerate the decomposition of some compounds.
  • the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
  • Anhydrous compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • anhydrous composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are generally packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
  • Oral dosage forms are either solid, gel or liquid.
  • the solid dosage forms are tablets, capsules, granules, and bulk powders.
  • Types of oral tablets include compressed, chewable lozenges and tablets which may be enteric-coated, sugar-coated or film-coated.
  • Capsules may be hard or soft gelatin capsules, while granules and powders may be provided in non-effervescent or effervescent form with the combination of other ingredients known to those skilled in the art.
  • the formulations are solid dosage forms such as for example, capsules or tablets.
  • the tablets, pills, capsules, troches and the like can contain one or more of the following ingredients, or compounds of a similar nature: a binder; a lubricant; a diluent; a glidant; a disintegrating agent; a coloring agent; a sweetening agent; a flavoring agent; a wetting agent; an enteric coating; a film coating agent and modified release agent.
  • binders include microcrystalline cellulose, methyl paraben, polyalkyleneoxides, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, molasses, polyvinylpyrrolidine, povidone, crospovidones, sucrose and starch and starch derivatives.
  • Lubricants include talc, starch, magnesium/calcium stearate, lycopodium and stearic acid.
  • Diluents include, for example, lactose, sucrose, trehalose, lysine, leucine, lecithin, starch, kaolin, salt, mannitol and dicalcium phosphate.
  • Glidants include, but are not limited to, colloidal silicon dioxide.
  • Disintegrating agents include crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose.
  • Coloring agents include, for example, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate and advanced coloring or anti-forgery color/opalescent additives known to those skilled in the art.
  • Sweetening agents include sucrose, lactose, mannitol and artificial sweetening agents such as saccharin, and any number of spray dried flavors.
  • Flavoring agents include natural flavors extracted from plants such as fruits and synthetic blends of compounds which produce a pleasant sensation or mask unpleasant taste, such as, but not limited to peppermint and methyl salicylate.
  • Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene laural ether.
  • Enteric-coatings include fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates.
  • Film coatings include hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.
  • Modified release agents include polymers such as the Eudragit® series and cellulose esters.
  • the compound, or derivative thereof can be provided in a composition that protects it from the acidic environment of the stomach.
  • the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine.
  • the composition may also be formulated in combination with an antacid or other such ingredient.
  • the dosage unit form When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil.
  • dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents.
  • the compounds can also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like.
  • a syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • the active materials can also be mixed with other active materials which do not impair the desired action, or with materials that supplement the desired action, such as antacids, H 2 blockers, and diuretics.
  • the active ingredient is a compound or derivative thereof as described herein. Higher concentrations, up to about 98% by weight of the active ingredient may be included.
  • tablets and capsules formulations may be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
  • they may be coated with a conventional enterically digestible coating, such as phenylsalicylate, waxes and cellulose acetate phthalate.
  • Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
  • Aqueous solutions include, for example, elixirs and syrups.
  • Emulsions are either oil-in-water or water-in-oil.
  • Elixirs are clear, sweetened, hydroalcoholic preparations.
  • Vehicles used in elixirs include solvents.
  • Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may contain a preservative.
  • An emulsion is a two-phase system in which one liquid is dispersed in the form of small globules throughout another liquid.
  • Carriers used in emulsions are non-aqueous liquids, emulsifying agents and preservatives.
  • Suspensions use suspending agents and preservatives.
  • Acceptable substances used in non-effervescent granules, to be reconstituted into a liquid oral dosage form include diluents, sweeteners and wetting agents.
  • Acceptable substances used in effervescent granules, to be reconstituted into a liquid oral dosage form include organic acids and a source of carbon dioxide. Coloring and flavoring agents are used in all of the above dosage forms.
  • Solvents include glycerin, sorbitol, ethyl alcohol and syrup.
  • preservatives include glycerin, methyl and propylparaben, benzoic acid, sodium benzoate and alcohol.
  • non-aqueous liquids utilized in emulsions include mineral oil and cottonseed oil.
  • emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate.
  • Suspending agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum and acacia.
  • Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as saccharin.
  • Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.
  • Organic acids include citric and tartaric acid.
  • Sources of carbon dioxide include sodium bicarbonate and sodium carbonate.
  • Coloring agents include any of the approved certified water soluble FD and C dyes, and mixtures thereof.
  • Flavoring agents include natural flavors extracted from plants such fruits, and synthetic blends of compounds which produce a pleasant taste sensation.
  • the solution or suspension in for example, propylene carbonate, vegetable oils or triglycerides, is in some embodiments encapsulated in a gelatin capsule.
  • a gelatin capsule Such solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545.
  • the solution e.g., for example, in a polyethylene glycol, may be diluted with a sufficient quantity of a liquid vehicle, e.g., water, to be easily measured for administration.
  • liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g., propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells.
  • Other useful formulations include those set forth in U.S. Pat. Nos. RE28,819 and 4,358,603.
  • such formulations include, but are not limited to, those containing a compound provided herein, a dialkylated mono- or polyalkylene glycol, including, but not limited to, 1,2-dimethoxyethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether wherein 350, 550 and 750 refer to the approximate average molecular weight of the polyethylene glycol, and one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, thiodipropionic acid and its esters, and dithiocarbamates.
  • BHT butylated
  • formulations include, but are not limited to, aqueous alcoholic solutions including a acetal.
  • Alcohols used in these formulations are any water-miscible solvents having one or more hydroxyl groups, including, but not limited to, propylene glycol and ethanol.
  • Acetals include, but are not limited to, di(lower alkyl) acetals of lower alkyl aldehydes such as acetaldehyde diethyl acetal.
  • injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • the injectables, solutions and emulsions also contain one or more excipients. Suitable excipients are, for example, water, saline, dextrose, glycerol or ethanol.
  • compositions to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.
  • a compound provided herein is dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene,
  • Parenteral administration of the compositions includes intravenous, subcutaneous and intramuscular administrations.
  • Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as lyophilized powders, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile emulsions.
  • the solutions may be either aqueous or nonaqueous.
  • suitable carriers include physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • PBS physiological saline or phosphate buffered saline
  • thickening and solubilizing agents such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • Vehicles used in parenteral preparations include aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other substances.
  • aqueous vehicles examples include Sodium Chloride Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection, Dextrose and Lactated Ringers Injection.
  • Nonaqueous parenteral vehicles include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil.
  • Antimicrobial agents in bacteriostatic or fungistatic concentrations must be added to parenteral preparations packaged in multiple-dose containers which include phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride.
  • Isotonic agents include sodium chloride and dextrose. Buffers include phosphate and citrate. Antioxidants include sodium bisulfate. Local anesthetics include procaine hydrochloride. Suspending and dispersing agents include sodium carboxymethylcelluose, hydroxypropyl methylcellulose and polyvinylpyrrolidone. Emulsifying agents include Polysorbate 80 (Tween® 80). A sequestering or chelating agent of metal ions includes EDTA. Carriers also include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles; and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.
  • the concentration of compound is adjusted so that an injection provides an effective amount to produce the desired pharmacological effect.
  • the exact dose depends on the age, weight, body surface area and condition of the patient or animal as is known in the art.
  • the unit-dose parenteral preparations are packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration must be sterile, as is known and practiced in the art.
  • intravenous or intraarterial infusion of a sterile aqueous solution containing an active compound is an effective mode of administration.
  • Another embodiment is a sterile aqueous or oily solution or suspension containing an active material injected as necessary to produce the desired pharmacological effect.
  • Injectables are designed for local and systemic administration.
  • a therapeutically effective dosage is formulated to contain a concentration of at least about 0.01% w/w up to about 90% w/w or more, in certain embodiments more than 0.1% w/w of the active compound to the treated tissue(s).
  • the compound may be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug.
  • the form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle.
  • the effective concentration is sufficient for ameliorating the symptoms of the condition and may be empirically determined.
  • Active ingredients provided herein can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,639,480; 5,733,566; 5,739,108; 5,891,474; 5,922,356; 5,972,891; 5,980,945; 5,993,855; 6,045,830; 6,087,324; 6,113,943; 6,197,350; 6,248,363; 6,264,970; 6,267,981; 6,376,461; 6,419,961; 6,589,548; 6,613,358; 6,699,500 and 6,740,634.
  • Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients provided herein.
  • controlled-release products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance.
  • controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release of other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time.
  • the drug In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body.
  • Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • the agent may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump may be used (see, Sefton, CRC Crit. Ref Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)).
  • polymeric materials can be used.
  • a controlled release system can be placed in proximity of the therapeutic target, i.e., thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical Applications of Controlled Release , vol. 2, pp. 115-138 (1984)).
  • a controlled release device is introduced into a subject in proximity of the site of inappropriate immune activation or a tumor. Other controlled release systems are discussed in the review by Langer ( Science 249:1527-1533 (1990)).
  • the active ingredient can be dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, ne
  • lyophilized powders which can be reconstituted for administration as solutions, emulsions and other mixtures. They may also be reconstituted and formulated as solids or gels.
  • the sterile, lyophilized powder is prepared by dissolving a compound provided herein, or a derivative thereof, in a suitable solvent.
  • the solvent may contain an excipient which improves the stability or other pharmacological component of the powder or reconstituted solution, prepared from the powder.
  • Excipients that may be used include, but are not limited to, an antioxidant, a buffer and a bulking agent.
  • the excipient is selected from dextrose, sorbital, fructose, corn syrup, xylitol, glycerin, glucose, sucrose and other suitable agent.
  • the solvent may contain a buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, at about neutral pH.
  • the resulting solution will be apportioned into vials for lyophilization.
  • Each vial will contain a single dosage or multiple dosages of the compound.
  • the lyophilized powder can be stored under appropriate conditions, such as at about 4° C. to room temperature.
  • Reconstitution of this lyophilized powder with water for injection provides a formulation for use in parenteral administration.
  • the lyophilized powder is added to sterile water or other suitable carrier. The precise amount depends upon the selected compound. Such amount can be empirically determined
  • Topical mixtures are prepared as described for the local and systemic administration.
  • the resulting mixture may be a solution, suspension, emulsions or the like and are formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical administration.
  • the compounds or derivatives thereof may be formulated as aerosols for topical application, such as by inhalation (see, e.g., U.S. Pat. Nos. 4,044,126, 4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment of inflammatory diseases, particularly asthma).
  • These formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microtine powder for insufflation, alone or in combination with an inert carrier such as lactose.
  • the particles of the formulation will, in some embodiments, have mass median geometric diameters of less than 5 microns, in other embodiments less than 10 microns.
  • Oral inhalation formulations of the compounds or derivatives suitable for inhalation include metered dose inhalers, dry powder inhalers and liquid preparations for administration from a nebulizer or metered dose liquid dispensing system.
  • metered dose inhalers dry powder inhalers
  • liquid preparations for administration from a nebulizer or metered dose liquid dispensing system for both metered dose inhalers and dry powder inhalers, a crystalline form of the compounds or derivatives is the preferred physical form of the drug to confer longer product stability.
  • crystalline particles of the compounds or derivatives can be generated using supercritical fluid processing which offers significant advantages in the production of such particles for inhalation delivery by producing respirable particles of the desired size in a single step.
  • a controlled particle size for the microcrystals can be selected to ensure that a significant fraction of the compounds or derivatives is deposited in the lung.
  • these particles have a mass median aerodynamic diameter of about 0.1 to about 10 microns, in other embodiments, about 1 to about 5 microns and still other embodiments, about 1.2 to about 3 microns.
  • HFA 134a (1,1,1,2-tetrafluoroethane) and HFA 227e (1,1,1,2,3,3,3-heptafluoropropane) and provided either alone or as a ratio to match the density of crystal particles of the compounds or derivatives.
  • a ratio is also selected to ensure that the product suspension avoids detrimental sedimentation or cream (which can precipitate irreversible agglomeration) and instead promote a loosely flocculated system, which is easily dispersed when shaken.
  • Loosely fluctuated systems are well regarded to provide optimal stability for pMDI canisters.
  • the formulation contained no ethanol and no surfactants/stabilizing agents.
  • TEMPO® MAP Pharmaceuticals, Inc., Mountain View, Calif.
  • pMDI pressurized metered dose inhalers
  • TEMPO® incorporates four novel features: 1) breath synchronous trigger—can be adjusted for different drugs and target populations to deliver the drug at a specific part of the inspiratory cycle, 2) plume control—an impinging jet to slow down the aerosol plume within the actuator, 3) vortexing chamber—consisting of porous wall, which provides an air cushion to keep the slowed aerosol plume suspended and air inlets on the back wall which drive the slowed aerosol plume into a vortex pattern, maintaining the aerosol in suspension and allowing the particle size to reduce as the HFA propellant evaporates, and 4) dose counter—will determine the doses remaining and prevent more than the intended maximum dose to be administered from any one canister.
  • the compounds may be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application.
  • Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies. Nasal solutions of the active compound alone or in combination with other excipients can also be administered.
  • the preparation may contain an esterified phosphonate compound dissolved or suspended in a liquid carrier, in particular, an aqueous carrier, for aerosol application.
  • a liquid carrier in particular, an aqueous carrier
  • the carrier may contain solubilizing or suspending agents such as propylene glycol, surfactants, absorption enhancers such as lecithin or cyclodextrin, or preservatives.
  • Solutions particularly those intended for ophthalmic use, may be formulated as 0.01%-10% isotonic solutions, pH about 5-7.4, with appropriate salts.
  • transdermal patches including iontophoretic and electrophoretic devices, and rectal administration, are also contemplated herein.
  • Transdermal patches including iotophoretic and electrophoretic devices, are well known to those of skill in the art.
  • such patches are disclosed in U.S. Pat. Nos. 6,267,983, 6,261,595, 6,256,533, 6,167,301, 6,024,975, 6,010715, 5,985,317, 5,983,134, 5,948,433 and 5,860,957.
  • dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect.
  • Rectal suppositories are used herein mean solid bodies for insertion into the rectum which melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients.
  • Substances utilized in rectal suppositories are bases or vehicles and agents to raise the melting point.
  • bases include cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol) and appropriate mixtures of mono-, di- and triglycerides of fatty acids. Combinations of the various bases may be used.
  • Agents to raise the melting point of suppositories include spermaceti and wax.
  • Rectal suppositories may be prepared either by the compressed method or by molding.
  • the weight of a rectal suppository in one embodiment, is about 2 to 3 gm.
  • Tablets and capsules for rectal administration are manufactured using the same substance and by the same methods as for formulations for oral administration.
  • the compounds provided herein, or derivatives thereof, may also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated. Many such targeting methods are well known to those of skill in the art. All such targeting methods are contemplated herein for use in the instant compositions. For non-limiting examples of targeting methods, see, e.g., U.S. Pat. Nos.
  • liposomal suspensions including tissue-targeted liposomes, such as tumor-targeted liposomes, may also be suitable as carriers. These may be prepared according to methods known to those skilled in the art. For example, liposome formulations may be prepared as described in U.S. Pat. No. 4,522,811. Briefly, liposomes such as multilamellar vesicles (MLV's) may be formed by drying down phosphatidyl choline and phosphatidyl serine (7:3 molar ratio) on the inside of a flask.
  • MLV's multilamellar vesicles
  • a solution of a compound provided herein in phosphate buffered saline lacking divalent cations (PBS) is added and the flask shaken until the lipid film is dispersed.
  • PBS phosphate buffered saline lacking divalent cations
  • the compounds or derivatives may be packaged as articles of manufacture containing packaging material, a compound or derivative thereof provided herein, which is effective for treatment, prevention or amelioration of one or more symptoms of the diseases or disorders, supra, within the packaging material, and a label that indicates that the compound or composition or derivative thereof, is used for the treatment, prevention or amelioration of one or more symptoms of the diseases or disorders, supra.
  • packaging materials for use in packaging products are well known to those of skill in the art. See, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252.
  • packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
  • a wide array of formulations of the compounds and compositions provided herein are contemplated as are a variety of treatments for any disease or disorder described herein.
  • compositions in other embodiments, should provide a dosage of from about 0.0001 mg to about 70 mg of compound per kilogram of body weight per day.
  • Dosage unit forms are prepared to provide from about 0.01 mg, 0.1 mg or 1 mg to about 500 mg, 1000 mg or 5000 mg, and in some embodiments from about 10 mg to about 500 mg of the active ingredient or a combination of essential ingredients per dosage unit form.
  • the amount of active ingredient in the formulations provided herein, which will be effective in the prevention or treatment of a disorder or one or more symptoms thereof, will vary with the nature and severity of the disease or condition, and the route by which the active ingredient is administered.
  • the frequency and dosage will also vary according to factors specific for each subject depending on the specific therapy (e.g., therapeutic or prophylactic agents) administered, the severity of the disorder, disease, or condition, the route of administration, as well as age, body, weight, response, and the past medical history of the subject.
  • Exemplary doses of a formulation include milligram or microgram amounts of the active compound per kilogram of subject (e.g., from about 1 micrograms per kilogram to about 50 milligrams per kilogram, from about 10 micrograms per kilogram to about 30 milligrams per kilogram, from about 100 micrograms per kilogram to about 10 milligrams per kilogram, or from about 100 microgram per kilogram to about 5 milligrams per kilogram).
  • compositions provided herein are also encompassed by the above described dosage amounts and dose frequency schedules.
  • the dosage administered to the subject may be increased to improve the prophylactic or therapeutic effect of the composition or it may be decreased to reduce one or more side effects that a particular subject is experiencing.
  • administration of the same formulation provided herein may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • the 5-HT 2A and 5-HT 2B activity agonist activity is lower than the 5-HT 2A and 5-HT 2B reported for cabergoline.
  • methods for antagonizing receptors including 5-HT 2A and 5-HT 2B receptors using the compounds and compositions, described herein. In practicing the methods, therapeutically effective amounts of the compounds or compositions, described herein, supra, are administered.
  • an ideal compound for such treatment should have selective agonist activities for the dopaminergic D 2 receptor.
  • the cabergoline-derivative compound hereinafter referred to as Compound 1,
  • Compound 1 exhibited weak serotonin 5-HT 2A and 5-HT 2B receptor agonist activity (as compared to cabergoline). Additionally, Compound 1 is selective for the dopaminergic receptor D 2L , compared to D 1 and D 4 receptors. Compound 1 also exhibited weak agonist activity on serotonin receptor 5-HT 1D . Compound 1 exhibited properties that are more desirable than cabergoline in that 5-HT 2A agonism is associated with hallucination, which is often a side effect of some anti-Parkinson's agents (see for example, Egan et al., Psychoparmacology (1998) 136:409-414). Agonist activity of the 5-HT 2B receptor has also been shown to be associated with cardiac and non-cardiac fibrosis.
  • the lower 5-HT 2B agonist activity seen with Compound 1 may translate into minimal or a reduction in the undesired fibrosis side-effects seen with cabergoline usage.
  • the compound may be advantageous for the compound to have a relatively short half-life. In some embodiments, the half-life of the compound is less than 5 hours. In other embodiments, the half-life of the compound is between 1 hour and 5 hours. In still other embodiments, the half-life of the compound is less than 8 hours. Alternatively, it may be advantageous for the compound to have a moderate to long half-life. In some embodiments, the half-life of the compound is greater than 10 hours. In other embodiments, the half-life of the compound is between 12 and 24 hours.
  • the compounds and compositions disclosed herein may also be used in combination with one or more other active ingredients.
  • the compounds may be administered in combination, or sequentially, with another therapeutic agent.
  • Such other therapeutic agents include, for example, those known for treatment, prevention, or amelioration of one or more symptoms associated with migraine or Parkinson's disease.
  • compounds such as Compound 1 may be administered to a patient with Parkinson's disease as an adjunct therapy in addition to monotherapies, such as L-dihydroxyphenylacetyl acid (L-DOPA).
  • L-DOPA L-dihydroxyphenylacetyl acid
  • compounds such as Compound 1 may be administered as a monotherapy.
  • any suitable combination of the compounds and compositions provided herein with one or more of the above therapeutic agents and optionally one or more further pharmacologically active substances are considered to be within the scope of the present disclosure.
  • the compounds and compositions provided herein are administered prior to or subsequent to the one or more additional active ingredients.
  • Compound 1 was synthesized as described above and assayed for receptor activity against a panel of dopaminergic and serotoninergic receptors.
  • Recombinant human serotonin receptors 5-HT 2A , and 5-HT 2B were expressed in CHO-K1 cells and activity (receptor agonist/antagonist activity) was assayed using an Aequorin assay (coexpression of mitochondrial apoaequorin).
  • Compound 1 was used at a concentration of 0.010 nM to 20,000 nM for agonist activity and 0.005 nM to 10,000 nM for antagonist activity. Results showed that Compound 1 displayed much less activity on 5-HT 2A and 5-HT 2B receptors as compared to cabergoline.
  • Compound 1 On 5-HT 2A receptors, Compound 1 had an EC 50 of 946 nM, compared to the reported EC 50 of 7.8 nM of cabergoline. On 5-HT 2B receptors, Compound 1 had an EC 50 of 6049 nM, compared to the reported EC50 of 2.6 nM of cabergoline (Newman-Tancredi, et al., J Pharmacology & Experimental Therapeutics (2002) 303(2): 815-822). The results from the receptor assays on human serotonin receptors indicate that Compound 1 may have a decreased or minimized occurrence of the undesired cardiac and non-cardiac fibrosis side-effects that are associated with cabergoline.
  • Recombinant human dopaminergic receptor D 1 , D 2L , and D 4 and serotonin receptor 5-HT 1B and 5-HT 1D activity was assayed using a GTP ⁇ S functional assay with Compound 1 at concentrations between 0.01 nM and 10,000 nM. Results showed that Compound 1 exhibited strong agonist activities with the D 2L receptor (EC 50 of 12.6 nM). Additionally, Compound 1 exhibited an 18-fold D 2L to D 4 receptor selectivity (when comparing EC 50 ). Compared to reported results for cabergoline, Compound 1 has a lower fold selectivity for D 2L than D 4 , cabergoline has approximately a 200-fold selectivity for the D 2L to D 4 receptor selectivity.
  • Compound 1 also exhibited a lower E max (26%) as compared to reported values for cabergoline (E max of 49%) (Newman-Tancredi, et al., J Pharmacology & Experimental Therapeutics (2002) 303(2): 805-814). Compound 1 was also highly selective for D 2L compared to D1 receptors, with results showing approximately 110-fold selectivity for the D 2L receptor over the D 1 receptor.

Abstract

Provided herein are novel cabergoline analogs and compositions thereof. In other embodiments, provided herein are methods of treatment, prevention, or amelioration of a variety of medical disorders such as, for example, Parkinson's disease using the compounds and compositions disclosed herein. In still other embodiments, provided herein are methods of antagonizing the 5-HT2B receptor and 5-HT2C receptors the using the compounds and compositions disclosed herein.

Description

    CROSS-REFERENCE
  • This application is a divisional application of U.S. patent application Ser. No. 13/531,432, filed on Jun. 22, 2012, the entire content of which is incorporated by reference.
  • FIELD
  • Provided herein are novel cabergoline analogs and compositions thereof. In other embodiments, provided herein are methods of treatment, prevention, or amelioration of a variety of medical disorders such as, for example, migraine and Parkinson's disease using the compounds and compositions disclosed herein. In some embodiments, provided herein are methods of agonizing dopaminergic D2 receptors and/or antagonizing or inhibiting activity of receptors such as the 5-HT2 serotonin receptors using the compounds and compositions disclosed herein. In other embodiments, provided herein are methods of agonizing receptors such as, for example, the 5-HT1D and/or the 5-HT1B receptor, without agonizing (or with very weak agonism of) the 5-HT2B receptor using the compounds and compositions disclosed herein.
  • BACKGROUND
  • Cabergoline, depicted below was approved for the treatment of Parkinson's disease but was subsequently withdrawn from the U.S. market because of undesirable agonization of the 5-HT2B receptor which leads to cardiac and non-cardiac fibrosis.
  • Figure US20150202198A1-20150723-C00001
  • Typically, an ideal anti-Parkinson's agent will be a selective agonist for dopaminergic D2 receptor. In some cases, it may be preferable to have a compound have a higher affinity (and stronger agonist activity) for the dopaminergic D2 receptor over the D1 and D4 receptors. Weak to modest agonist activity on the 5-HT1B and/or 5-HT1D receptors may also be desirable. Studies have also shown that 5-HT2 receptor antagonism may also be desirable for the treatment of Parkinson's disease symptoms and/or for the reduction of undesirable side-effects, such as cardiac or non-cardiac fibrosis and psychiatric side-effects (Newman-Trancredi, J Pharmacology and Experimental Therapies (2002) 303(2):815-822). Depending on the treatment regimen, anti-Parkinson's agents may have a relatively short half-life (i.e., 1-5 hours) or a longer half-life (i.e., 20 hours or longer).
  • Accordingly, what is needed are cabergoline analogs which retain activity against central nervous disorders, such as, for example, Parkinson's disease and lack agonist activity, or very weak agonist activity, against the 5-HT2 receptors, including 5-HT2B and the 5-HT2C receptors.
  • SUMMARY
  • Provided herein are cabergoline derivatives which address these and other needs. In one aspect, cabergoline derivatives described herein include compounds of Formula (I) or (II):
  • Figure US20150202198A1-20150723-C00002
  • or ion pairs, salts, hydrates, solvates or metabolites thereof, wherein:
  • R1 is hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms;
  • R2 is alkyl, substituted alkyl, acyl, substituted acyl, halo, heteroalkyl, substituted heteroalkyl, —NO2, —N3, —OH, —S(O)kR100, —OR101, —NR102R103, —CONR104R105, —CO2R106 or —O2CR107;
  • R5 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, arylalkyl or substituted arylalkyl;
  • R7 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, arylalkyl or substituted arylalkyl;
  • R8 and R9 are independently hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, heteroalkyl, substituted heteroalkyl, arylalkyl, substituted arylalkyl, heteroaryl, substituted heteroaryl or R4 and R5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring
  • R11 is hydrogen, (C1-C3) alkyl or (C1-C3) alkyl substituted with one or more fluorine atoms;
  • R100-R107 are independently hydrogen, alkyl, substituted alkyl, acyl, substituted acyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl or substituted heteroarylalkyl;
  • k is 0, 1 or 2; and
  • n is 0, 1, 2 or 3.
  • Also provided are derivatives, including salts, esters, enol ethers, enol esters, solvates, hydrates and prodrugs of the compounds described herein. Further provided are compositions which include the compounds provided herein and a vehicle.
  • Methods of treating, preventing, or ameliorating symptoms of medical disorders such as, for example, central nervous system disorders, migraine and Parkinson's disease are also provided herein. In practicing the methods, therapeutically effective amounts of the compounds or compositions thereof are administered to a subject.
  • Methods of antagonizing receptors such as, for example, the 5-HT2A and 5-HT2B with the compounds and compositions described herein are also provided herein. Methods of agonizing dopaminergic D2 receptors using the compounds and compositions disclosed herein are also provided. In practicing the methods, therapeutically effective amounts of the compounds or compositions are administered.
  • In some embodiments, methods for selectively agonizing dopaminergic D2 receptors (over other dopaminergic receptors) are provided. In other embodiments, methods of antagonizing or inhibiting (or not agonizing) activity of the 5-HT2 receptors are also provided. In other embodiments methods and compounds for both selectively agonizing dopaminergic D2 receptors and antagonizing (or not agonizing/very weak agonist activity) 5-HT2 receptors at the same time are provided. Such methods and compounds may be desirable as agents for treating one or more symptoms of Parkinson's disease.
  • DETAILED DESCRIPTION Definitions
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this invention belongs. In the event that there is a plurality of definitions for a term herein, those in this section prevail unless stated otherwise.
  • “Alkyl,” by itself or as part of another substituent, refers to a saturated or unsaturated, branched, straight-chain or cyclic monovalent hydrocarbon radical derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane, alkene or alkyne. Typical alkyl groups include, but are not limited to, methyl; ethyls such as ethanyl, ethenyl, ethynyl; propyls such as propan-1-yl, propan-2-yl, cyclopropan-1-yl, prop-1-en-1-yl, prop-1-en-2-yl, prop-2-en-1-yl (allyl), cycloprop-1-en-1-yl; cycloprop-2-en-1-yl, prop-1-yn-1-yl, prop-2-yn-1-yl, etc.; butyls such as butan-1-yl, butan-2-yl, 2-methyl-propan-1-yl, 2-methyl-propan-2-yl, cyclobutan-1-yl, but-1-en-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-1-yl, but-2-en-2-yl, buta-1,3-dien-1-yl, buta-1,3-dien-2-yl, cyclobut-1-en-1-yl, cyclobut-1-en-3-yl, cyclobuta-1,3-dien-1-yl, but-1-yn-1-yl, but-1-yn-3-yl, but-3-yn-1-yl, etc.; and the like. The term “alkyl” is specifically intended to include groups having any degree or level of saturation, i.e., groups having exclusively single carbon-carbon bonds, groups having one or more double carbon-carbon bonds, groups having one or more triple carbon-carbon bonds and groups having mixtures of single, double and triple carbon-carbon bonds. Where a specific level of saturation is intended, the expressions “alkanyl,” “alkenyl,” and “alkynyl” are used. In some embodiments, an alkyl group comprises from 1 to 20 carbon atoms (C1-C20 alkyl). In other embodiments, an alkyl group comprises from 1 to 10 carbon atoms (C1-C10 alkyl). In still other embodiments, an alkyl group comprises from 1 to 6 carbon atoms (C1-C6 alkyl).
  • “Alkanyl,” by itself or as part of another substituent, refers to a saturated branched, straight-chain or cyclic alkyl radical derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane. Typical alkanyl groups include, but are not limited to, methanyl; ethanyl; propanyls such as propan-1-yl, propan-2-yl (isopropyl), cyclopropan-1-yl, etc.; butanyls such as butan-1-yl, butan-2-yl (sec-butyl), 2-methyl-propan-1-yl (isobutyl), 2-methyl-propan-2-yl (t-butyl), cyclobutan-1-yl, etc.; and the like.
  • “Alkenyl,” by itself or as part of another substituent, refers to an unsaturated branched, straight-chain or cyclic alkyl radical having at least one carbon-carbon double bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkene. The group may be in either the cis or trans conformation about the double bond(s). Typical alkenyl groups include, but are not limited to, ethenyl; propenyls such as prop-1-en-1-yl, prop-1-en-2-yl, prop-2-en-1-yl (allyl), prop-2-en-2-yl, cycloprop-1-en-1-yl; cycloprop-2-en-1-yl; butenyls such as but-1-en-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-1-yl, but-2-en-1-yl, but-2-en-2-yl, buta-1,3-dien-1-yl, buta-1,3-dien-2-yl, cyclobut-1-en-1-yl, cyclobut-1-en-3-yl, cyclobuta-1,3-dien-1-yl, etc.; and the like.
  • “Alkynyl,” by itself or as part of another substituent refers to an unsaturated branched, straight-chain or cyclic alkyl radical having at least one carbon-carbon triple bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkyne. Typical alkynyl groups include, but are not limited to, ethynyl; propynyls such as prop-1-yn-1-yl, prop-2-yn-1-yl, etc.; butynyls such as but-1-yn-1-yl, but-1-yn-3-yl, but-3-yn-1-yl, etc.; and the like.
  • “Acyl” by itself or as part of another substituent refers to a radical —C(O)R400, where R400 is hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroalkyl, substituted heteroalkyl, heteroarylalkyl or substituted heteroarylalkyl as defined herein. Representative examples include, but are not limited to formyl, acetyl, cyclohexylcarbonyl, cyclohexylmethylcarbonyl, benzoyl, benzylcarbonyl and the like.
  • “Aryl,” by itself or as part of another substituent, refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system, as defined herein. Typical aryl groups include, but are not limited to, groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene and the like. In some embodiments, an aryl group comprises from 6 to 20 carbon atoms (C6-C20 aryl). In other embodiments, an aryl group comprises from 6 to 15 carbon atoms (C6-C15 aryl). In still other embodiments, an aryl group comprises from 6 to 15 carbon atoms (C6-C10 aryl).
  • “Arylalkyl,” by itself or as part of another substituent, refers to an acyclic alkyl group in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl group as, as defined herein. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, 2-phenylethen-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, 2-naphthylethen-1-yl, naphthobenzyl, 2-naphthophenylethan-1-yl and the like. Where specific alkyl moieties are intended, the nomenclature arylalkanyl, arylalkenyl and/or arylalkynyl is used. In some embodiments, an arylalkyl group is (C6-C30) arylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is (C1-C10) alkyl and the aryl moiety is (C6-C20) aryl. In other embodiments, an arylalkyl group is (C6-C20) arylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is (C1-C8) alkyl and the aryl moiety is (C6-C12) aryl. In still other embodiments, an arylalkyl group is (C6-C15) arylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the arylalkyl group is (C1-C5) alkyl and the aryl moiety is (C6-C10) aryl.
  • “Compounds” refers to compounds encompassed by structural formulae disclosed herein and includes any specific compounds within these formulae whose structure is disclosed herein. Compounds may be identified either by their chemical structure and/or chemical name. When the chemical structure and chemical name conflict, the chemical structure is determinative of the identity of the compound. The compounds described herein may contain one or more chiral centers and/or double bonds and therefore, may exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), enantiomers or diastereomers. Accordingly, the chemical structures depicted herein encompass all possible enantiomers and stereoisomers of the illustrated compounds including the stereoisomerically pure form (e.g., geometrically pure, enantiomerically pure or diastereomerically pure) and enantiomeric and stereoisomeric mixtures. Enantiomeric and stereoisomeric mixtures can be resolved into their component enantiomers or stereoisomers using separation techniques or chiral synthesis techniques well known to the skilled artisan. The compounds may also exist in several tautomeric forms including the enol form, the keto form and mixtures thereof. Accordingly, the chemical structures depicted herein encompass all possible tautomeric forms of the illustrated compounds. The compounds described also include isotopically labeled compounds where one or more atoms have an atomic mass different from the atomic mass conventionally found in nature. Examples of isotopes that may be incorporated into the compounds described herein include, but are not limited to, 2H, 3H, 13C, 14C, 15N, 18O, 17O, 35S, etc. In general, it should be understood that all isotopes of any of the elements comprising the compounds described herein may be found in these compounds. Compounds may exist in unsolvated or unhydrated forms as well as solvated forms, including hydrated forms and as N-oxides. In general, compounds may be hydrated, solvated or N-oxides. Certain compounds may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated herein and are intended to be within the scope of the present invention. Further, it should be understood, when partial structures of the compounds are illustrated, that brackets indicate the point of attachment of the partial structure to the rest of the molecule.
  • “Heteroalkyl,” “Heteroalkanyl,” “Heteroalkenyl” and “Heteroalkynyl,” by themselves or as part of other substituents, refer to alkyl, alkanyl, alkenyl and alkynyl groups, respectively, in which one or more of the carbon atoms (and optionally any associated hydrogen atoms), are each, independently of one another, replaced with the same or different heteroatoms or heteroatomic groups. Typical heteroatoms or heteroatomic groups which can replace the carbon atoms include, but are not limited to, —O—, —S—, —N—, —Si—, —NH—, —S(O)—, —S(O)2—, —S(O)NH—, —S(O)2NH— and the like and combinations thereof. The heteroatoms or heteroatomic groups may be placed at any interior position of the alkyl, alkenyl or alkynyl groups. Typical heteroatomic groups which can be included in these groups include, but are not limited to, —O—, —S—, —O—O—, —S—S—, —O—S—, —NR501R502—, ═N—N═, —N═N—, —N═N—NR503R404, —PR505—, —P(O)2—, —POR506—, —O—P(O)2—, —SO—, —SO2—, —SnR507R508— and the like, where R501, R502, R503, R504, R505, R506, R507 and R508— are independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, cycloalkyl, substituted cycloalkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl or substituted heteroarylalkyl.
  • “Heteroaryl,” by itself or as part of another substituent, refers to a monovalent heteroaromatic radical derived by the removal of one hydrogen atom from a single atom of a parent heteroaromatic ring systems, as defined herein. Typical heteroaryl groups include, but are not limited to, groups derived from acridine, β-carboline, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine, quinazoline, quinoline, quinolizine, quinoxaline, tetrazole, thiadiazole, thiazole, thiophene, triazole, xanthene, and the like. In some embodiments, the heteroaryl group comprises from 5 to 20 ring atoms (5-20 membered heteroaryl). In other embodiments, the heteroaryl group comprises from 5 to 10 ring atoms (5-10 membered heteroaryl). Exemplary heteroaryl groups include those derived from furan, thiophene, pyrrole, benzothiophene, benzofuran, benzimidazole, indole, pyridine, pyrazole, quinoline, imidazole, oxazole, isoxazole and pyrazine.
  • “Heteroarylalkyl” by itself or as part of another substituent refers to an acyclic alkyl group in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heteroaryl group. Where specific alkyl moieties are intended, the nomenclature heteroarylalkanyl, heteroarylakenyl and/or heteroarylalkynyl is used. In some embodiments, the heteroarylalkyl group is a 6-21 membered heteroarylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety of the heteroarylalkyl is (C1-C6) alkyl and the heteroaryl moiety is a 5-15-membered heteroaryl. In other embodiments, the heteroarylalkyl is a 6-13 membered heteroarylalkyl, e.g., the alkanyl, alkenyl or alkynyl moiety is (C1-C3) alkyl and the heteroaryl moiety is a 5-10 membered heteroaryl.
  • “Hydrates” refers to incorporation of water into to the crystal lattice of a compound described herein, in stochiometric proportions, resulting in the formation of an adduct. Methods of making hydrates include, but are not limited to, storage in an atmosphere containing water vapor, dosage forms that include water, or routine pharmaceutical processing steps such as, for example, crystallization (i.e., from water or mixed aqueous solvents), lyophilization, wet granulation, aqueous film coating, or spray drying. Hydrates may also be formed, under certain circumstances, from crystalline solvates upon exposure to water vapor, or upon suspension of the anhydrous material in water. Hydrates may also crystallize in more than one form resulting in hydrate polymorphism. See e.g., (Guillory, K., Chapter 5, pp. 202-205 in Polymorphism in Pharmaceutical Solids, (Brittain, H. ed.), Marcel Dekker, Inc., New York, N.Y., 1999). The above methods for preparing hydrates are well within the ambit of those of skill in the art, are completely conventional and do not require any experimentation beyond what is typical in the art. Hydrates may be characterized and/or analyzed by methods well known to those of skill in the art such as, for example, single crystal X-Ray diffraction, X-Ray powder diffraction, Polarizing optical microscopy, thermal microscopy, thermogravimetry, differential thermal analysis, differential scanning calorimetry, IR spectroscopy, Raman spectroscopy and NMR spectroscopy. (Brittain, H., Chapter 6, pp. 205-208 in Polymorphism in Pharmaceutical Solids, (Brittain, H. ed.), Marcel Dekker, Inc. New York, 1999). In addition, many commercial companies routine offer services that include preparation and/or characterization of hydrates such as, for example, HOLODIAG, Pharmaparc II, Voie de l'Innovation, 27 100 Val de Reuil, France (http://www.holodiag.com).
  • “Parent Aromatic Ring System” refers to an unsaturated cyclic or polycyclic ring system having a conjugated π electron system. Specifically included within the definition of “parent aromatic ring system” are fused ring systems in which one or more of the rings are aromatic and one or more of the rings are saturated or unsaturated, such as, for example, fluorene, indane, indene, phenalene, etc. Typical parent aromatic ring systems include, but are not limited to, accanthrylene, accnaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene and the like.
  • “Preventing” or “prevention” refers to a reduction in risk of acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a patient that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease). In some embodiments, “preventing” or “prevention” refers to reducing symptoms of the disease by taking the compound in a preventative fashion. The application of a therapeutic for preventing or prevention of a disease of disorder is known as ‘prophylaxis.’ In some embodiments, the compounds provided herein provide superior prophylaxis because of lower long term side effects over long time periods.
  • “Prodrug” refers to a derivative of a drug molecule that requires a transformation within the body to release the active drug. Prodrugs are frequently (though not necessarily) pharmacologically inactive until converted to the parent drug.
  • “Promoiety” refers to a form of protecting group that when used to mask a functional group within a drug molecule converts the drug into a prodrug. Typically, the promoiety will be attached to the drug via bond(s) that are cleaved by enzymatic or non-enzymatic means in vivo.
  • “Salt” refers to a salt of a compound, which possesses the desired pharmacological activity of the parent compound. Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine and the like. In some embodiments, the salt is pharmaceutically acceptable.
  • “Solvates” refers to incorporation of solvents into to the crystal lattice of a compound described herein, in stochiometric proportions, resulting in the formation of an adduct. Methods of making solvates include, but are not limited to, storage in an atmosphere containing a solvent, dosage forms that include the solvent, or routine pharmaceutical processing steps such as, for example, crystallization (i.e., from solvent or mixed solvents) vapor diffusion, etc. Solvates may also be formed, under certain circumstances, from other crystalline solvates or hydrates upon exposure to the solvent or upon suspension material in solvent. Solvates may crystallize in more than one form resulting in solvate polymorphism. See e.g., (Guillory, K., Chapter 5, pp. 205-208 in Polymorphism in Pharmaceutical Solids, (Brittain, H. ed.), Marcel Dekker, Inc., New York, N.Y., 1999)). The above methods for preparing solvates are well within the ambit of those of skill in the art, are completely conventional do not require any experimentation beyond what is typical in the art. Solvates may be characterized and/or analyzed by methods well known to those of skill in the art such as, for example, single crystal X-Ray diffraction, X-Ray powder diffraction, Polarizing optical microscopy, thermal microscopy, thermogravimetry, differential thermal analysis, differential scanning calorimetry, IR spectroscopy, Raman spectroscopy and NMR spectroscopy. (Brittain, H., Chapter 6, pp. 205-208 in Polymorphism in Pharmaceutical Solids, (Brittain, H. ed.), Marcel Dekker, Inc. New York, 1999). In addition, many commercial companies routine offer services that include preparation and/or characterization of solvates such as, for example, HOLODIAG, Pharmaparc II, Voie de l'Innovation, 27 100 Val de Reuil, France (http://wwwholodiag.com).
  • “Substituted,” when used to modify a specified group or radical, means that one or more hydrogen atoms of the specified group or radical are each, independently of one another, replaced with the same or different substituent(s). Substituent groups useful for substituting saturated carbon atoms in the specified group or radical include, but are not limited to —Ra, halo, —O, ═ORb, —SRb, —S, ═S, —NRcRc, ═NRb, ═N—ORb, trihalomethyl, —CF3, —CN, —OCN, —SCN, —NO, —NO2, ═N2, —N3, —S(O)2Rb, —S(O)2NRb, —S(O)2O, —S(O)2ORb, —OS(O)2Rb, —OS(O)2O, —OS(O)2ORb, —P(O)(O)2, —P(O)(ORb)(O), —P(O)(ORb)(ORb), —C(O)Rb, —C(S)Rb, —C(NRb)Rb, —C(O)O, —C(O)ORb, —C(S)ORb, —C(O)NRcRc, —C(NRb)NRcRc, —OC(O)Rb, —OC(S)Rb, —OC(O)O, —OC(O)ORb, —OC(S)ORb, —NRbC(O)Rb, —NRbC(S)Rb, —NRbC(O)O, —NRbC(O)ORb, —NRbC(S)ORb, —NRbC(O)NRcRc, —NRbC(NRb)Rb and —NRbC(NRb)NRcRc, where Ra is selected from the group consisting of alkyl, cycloalkyl, heteroalkyl, cycloheteroalkyl, aryl, arylalkyl, heteroaryl and heteroarylalkyl; each Rb is independently hydrogen or Ra; and each Rc is independently Rb or alternatively, the two Rcs are taken together with the nitrogen atom to which they are bonded form a 4-, 5-, 6- or 7-membered cycloheteroalkyl which may optionally include from 1 to 4 of the same or different additional heteroatoms selected from the group consisting of O, N and S. As specific examples, —NRcRc is meant to include —NH2, —NH-alkyl, N-pyrrolidinyl and N-morpholinyl.
  • Similarly, substituent groups useful for substituting unsaturated carbon atoms in the specified group or radical include, but are not limited to, —Ra, halo, —O, —ORb, —SRb, —S, —NRcRc, trihalomethyl, —CF3, —CN, —OCN, —SCN, —NO, —NO2, —N3, —S(O)2Rb, —S(O)2O, —S(O)2ORb, —OS(O)2Rb, —OS(O)2O, —OS(O)2ORb, —P(O)(O)2, —P(O)(ORb)(O), —P(O)(ORb)(ORb), —C(O)Rb, —C(S)Rb, —C(NRb)Rb, —C(O)O, —C(O)ORb, —C(S)ORb, —C(O)NRcRc, —C(NRb)NRcRc, —OC(O)Rb, —OC(S)Rb, —OC(O)O, —OC(O)ORb, —OC(S)ORb, —NRbC(O)Rb, —NRbC(S)Rb, —NRbC(O)O, —NRbC(O)ORb, —NRbC(S)ORb, —NRbC(O)NRcRc, —NRbC(NRb)Rb and —NRbC(NRb)NRcRc, where Ra, Rb and Rc are as previously defined.
  • Substituent groups useful for substituting nitrogen atoms in heteroalkyl and cycloheteroalkyl groups include, but are not limited to, —Ra, —O, —ORb, —SRb, —S, —NRcRc, trihalomethyl, —CF3, —CN, —NO, —NO2, —S(O)2Rb, —S(O)2O, —S(O)2ORb, —OS(O)2Rb, —OS(O)2O, —OS(O)2ORb, —P(O)(O)2, —P(O)(ORb)(O), —P(O)(ORb)(ORb), —C(O)Rb, —C(S)Rb, —C(NRb)Rb, —C(O)ORb, —C(S)ORb, —C(O)NRcRc, —C(NRb)NRcRc, —OC(O)Rb, —OC(S)Rb, —OC(O)OR b, —OC(S)ORb, —NRbC(O)Rb, —NRbC(S)Rb, —NRbC(O)ORb, —NRbC(S)ORb, —NRbC(O)NRcRc, —NRbC(NRb)Rb and —NRbC(NRb)NRcRc, where Ra, Rb and Rc are as previously defined.
  • Substituent groups from the above lists useful for substituting other specified groups or atoms will be apparent to those of skill in the art. The substituents used to substitute a specified group can be further substituted, typically with one or more of the same or different groups selected from the various groups specified above. In some embodiments, substituents are limited to the groups above.
  • “Subject,” “individual” or “patient” is used interchangeably herein and refers to a vertebrate, preferably a mammal Mammals include, but are not limited to, murines, rodents, simians, humans, farm animals, sport animals and pets.
  • “Treating” or “treatment” of any disease or disorder refers, in some embodiments, to ameliorating the disease or disorder (i.e., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof,). Treatment can also refer to the lessening the severity and/or the duration of one or more symptoms of a disease or disorder. Treatment may also be considered to include preemptive or prophylactic administration to ameliorate, arrest or prevent the development of the disease or at least one of the clinical symptoms. Treatment can also refer to the lessening of the severity and/or the duration of one or more symptoms of a disease or disorder. In a further feature, the treatment rendered has lower potential for long term side effects over multiple years. In other embodiments “treating” or “treatment” refers to ameliorating at least one physical parameter, which may not be discernible by the patient. In yet other embodiments, “treating” or “treatment” refers to inhibiting the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter) or both. In yet other embodiments, “treating” or “treatment” refers to delaying the onset of the disease or disorder.
  • “Therapeutically effective amount” means the amount of a compound that, when administered to a patient for treating a disease, is sufficient to effect such treatment for the disease. The “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, adsorption, distribution, metabolism and excretion etc., of the patient to be treated.
  • “Vehicle” refers to a diluent, excipient or carrier with which a compound is administered to a subject. In some embodiments, the vehicle is pharmaceutically acceptable.
  • Compounds
  • Provided herein are compounds of Formula (I) or (II):
  • Figure US20150202198A1-20150723-C00003
  • or ion pairs, salts, hydrates, solvates or metabolites thereof, wherein:
  • R1 is hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms;
  • R2 is alkyl, substituted alkyl, acyl, substituted acyl, halo, heteroalkyl, substituted heteroalkyl, —NO2, —N3, —OH, —S(O)kR100, —OR101 , —NR102R103, —CONR104R105, —CO2R106 or —O2CR107;
  • R5 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, arylalkyl or substituted arylalkyl;
  • R7 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, arylalkyl or substituted arylalkyl;
  • R8 and R9 are independently hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, heteroalkyl, substituted heteroalkyl, arylalkyl, substituted arylalkyl, heteroaryl, substituted heteroaryl or R4 and R5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring
  • R11 is hydrogen, (C1-C3) alkyl or (C1-C3) alkyl substituted with one or more fluorine atoms;
  • R100-R107 are independently hydrogen, alkyl, substituted alkyl, acyl, substituted acyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl or substituted heteroarylalkyl;
  • k is 0, 1 or 2; and
  • n is 0, 1, 2 or 3.
  • In some embodiments, R1 and R11 are not both hydrogen. In other embodiments, when R11 is hydrogen, R1 is substituted (C1-C4) alkyl with one or more fluorine atoms. In still other embodiments, when R11 is hydrogen, R1 is (C1-C4) alkyl substituted with one or more fluorine atoms. In still other embodiments, when R1 is (C1-C4) alkyl substituted with one or more fluorine atoms. In still other embodiments, R11 is (C1-C3) alkyl substituted with one or more fluorine atoms. In still other embodiments, R11is —CF3.
  • In some embodiments, R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms. In other embodiments, R1 is hydrogen, —CH3 or —CF3.
  • In some embodiments, R2 is alkyl, acyl, halo, —NO2, —OH, —S(O)kR100, —OR101, —NR102R103, —CONR104R105, —CO2R106 or —O2CR107. In other embodiments, R2 is alkyl, acyl, halo, —NO2, —OH, —S(O)kR100, —OR101, —NR102R103, —CONR104R105, —CO2R106 or —O2CR107 and n is 1. In still other embodiments, R2 is alkyl, halo or —OR101 and n is 1. In still other embodiments, n is 0.
  • In some embodiments, R5 is hydrogen or (C1-C3) alkyl. In other embodiments, R5 is hydrogen, methyl or allyl. In still other embodiments, R5 is methyl. In still other embodiments, R5 is allyl.
  • In some embodiments, R7 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms. In other embodiments, R7 is hydrogen or (C1-C4) alkyl substituted with one dialkyl amino group. In still other embodiments, R8 and R9 are independently hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, or R4 and R5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring.
  • In some embodiments, R8 and R9 are independently hydrogen or (C1-C4) alkyl. In other embodiments, R8 is hydrogen and R9 is (C1-C4) alkyl.
  • In some embodiments, R11 is (C1-C3) alkyl substituted with one or more fluorine atoms. In other embodiments, R11 is —CF3.
  • In some embodiments, R100-R122 are independently hydrogen, alkyl, or substituted alkyl, acyl or substituted acyl. In some embodiments, R100-R122 are independently hydrogen or alkyl.
  • In some embodiments, R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R2 is alkyl, acyl, halo, —NO2, —OH, —S(O)kR100, —OR101, —NR102R103, —CONR104R105, —CO2R106 or —O2CR107 and n is 1, R5 is hydrogen or (C1-C3) alkyl, R7 is hydrogen, (C1-C4) alkyl or (C1-C4) substituted alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, R8 and R9 are independently hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, or R4 and R5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring and R11 is (C1-C3) alkyl substituted with one or more fluorine atoms. In other embodiments, R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, n is 0, R5 is hydrogen or (C1-C3) alkyl, R7 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, R8 and R9 are independently hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, or R4 and R5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring and R11 is (C1-C3) alkyl substituted with one or more fluorine atoms.
  • In some embodiments, R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R2 is alkyl, acyl, halo, —NO2, —OH, —S(O)kR100, —OR100, —NR102R103, —CONR104R105, —CO2R106 or —O2CR107 and n is 1, fluoro, hydroxy or methoxy, R5 is hydrogen or (C1-C3) alkyl, R7 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R8 and R9 are independently hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, or R4 and R5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring and R11 is (C1-C3) alkyl substituted with one or more fluorine atoms. In other embodiments, R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, n is 0, R5 is hydrogen or (C1-C3) alkyl, R7 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R8 and R9 are independently hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl, (C1-C4) alkyl substituted with one or more fluorine atoms, or R4 and R5 together with the nitrogen atom to which they are attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring and R11 is (C1-C3) alkyl substituted with one or more fluorine atoms.
  • In some embodiments, R1 is hydrogen, —CH3 or —CF3, R2 is alkyl, halo or —OR101 and n is 1, R5 is hydrogen, methyl or allyl, R7 is hydrogen or (C1-C4) alkyl substituted with one dimethyl amino group, R8 and R9 are independently hydrogen, (C1-C4) alkyl or (C1-C4) substituted alkyl and R11 is —CF3. In other embodiments, R1 is hydrogen, —CH3 or —CF3, n is 0, R5 is hydrogen, methyl or allyl, R7 is hydrogen or (C1-C4) alkyl substituted with one dimethyl amino group, R8 and R9 are independently hydrogen, (C1-C4) alkyl or (C1-C4) substituted alkyl and R11 is —CF3.
  • In some embodiments, R1 is hydrogen, —CH3 or —CF3, R2 is alkyl, halo or —OR101 and n is 1, R5 is hydrogen, methyl or allyl, R7 is hydrogen or (C1-C4) alkyl substituted with one dimethyl amino group, R8 and R9 are independently hydrogen, (C1-C4) alkyl or (C1-C4) substituted alkyl and R11 is —CF3. In other embodiments, R1 is hydrogen, —CH3 or —CF3, n is 0, R5 is hydrogen, methyl or allyl, R7 is hydrogen or (C1-C4) alkyl substituted with one dimethyl amino group, R8 and R9 are independently hydrogen, (C1-C4) alkyl or (C1-C4) substituted alkyl and R11 is —CF3.
  • In some embodiments a compound of Formula (III) or (IV):
  • Figure US20150202198A1-20150723-C00004
  • is provided.
  • In some embodiments, R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms. In other embodiments, R1 is hydrogen, —CH3 or —CF3. In still other embodiments, R1 is —CH3 or —CF3,
  • In some embodiments, R5 is hydrogen or (C1-C3) alkyl. In other embodiments, R5 is hydrogen, methyl or allyl. In still other embodiments, R5 is methyl or allyl.
  • In some embodiments, R2 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms. In other embodiments, R7 is substituted (C1-C4) alkyl or (C1-C4) alkyl substituted with one dialkyl amino group. In till other embodiments, R7 is 3-dimethylaminopropyl.
  • In some embodiments, R9 is hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms. In other embodiments, R9 is (C1-C4) alkyl or (C1-C4) substituted alkyl. In still other embodiments, R9 is (C1-C4) alkyl. In still other embodiments, R9 is ethyl.
  • In some embodiments, R11 is hydrogen or (C1-C3) alkyl substituted with one or more fluorine atoms. In other embodiments, R11 is —CF3.
  • In some embodiments, R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R5 is hydrogen or (C1-C3) alkyl, R7 is hydrogen, (C1-C4) alkyl, (C1-C4) substituted alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R9 is hydrogen, (C1-C4) alkyl, substituted (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms and R11 is hydrogen or (C1-C3) alkyl substituted with one or more fluorine atoms. In other embodiments, R1 is hydrogen, —CH3 or —CF3, R5 is hydrogen, methyl or allyl, R7 is substituted (C1-C4) alkyl or (C1-C4) alkyl substituted with one dialkyl amino group, R9 is (C1-C4) alkyl and R11 is —CF3. In still other embodiments, R1 is —CH3 or —CF3, R5 is methyl or allyl, R7 (C1-C4) alkyl substituted with one dialkyl amino group R9 is (C1-C4) alkyl and R11 is —CF3.
  • In some embodiments, compounds of structure (V) and (VI):
  • Figure US20150202198A1-20150723-C00005
  • are provided wherein R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R5 is hydrogen or (C1-C3) alkyl, and R11 is (C1-C3) alkyl substituted with one or more fluorine atoms.
  • In some embodiments, R1 is hydrogen, —CH3 or —CF3, R5 is hydrogen, methyl or allyl, and R11 is hydrogen or —CF3. In other embodiments, R1 is hydrogen, R5 is allyl, and R11 is —CF3. In still other embodiments, R1 is —CH3 or —CF3, R5 is methyl or allyl, and R11 is —CF3. In still other embodiments, R1 is —CH3 or —CF3, R5 is methyl or allyl, and R11 is —CF3.
  • Exemplary compounds of Formula (I) include the compounds depicted below.
  • Figure US20150202198A1-20150723-C00006
    Figure US20150202198A1-20150723-C00007
    Figure US20150202198A1-20150723-C00008
  • Exemplary compounds of Formula (II) include, for example, the 8,9-dihydro analogues of the above depicted compounds of Formula (I). In some embodiments, R1 is hydrogen, —CH3 or —CF3, and R11 is hydrogen or —CF3. In other embodiments, R1 is —CH3 or —CF3 and R11 is —CF3.
  • Exemplary methods for the preparation of compounds of Formula (I) and (II) for use in the compositions and methods provided herein are described below and in the Examples but other methods known in the art can be used to prepare the cabergoline derivatives disclosed herein.
  • In some embodiments, direct functionalization of 2-unsubstituted analogs of compounds of Formula (I) and (II) (e.g., compounds of Formula (VII) and (VIII)), for example, with an alkyl halide under basic conditions can be used to provide the compounds of Formula (I) and (II).
  • Figure US20150202198A1-20150723-C00009
  • In other embodiments, acids (IX) and (X) which can be prepared by methods well known to those of skill in the art can be used provide compounds of Formulas (I) and (II).
  • Figure US20150202198A1-20150723-C00010
  • Many methods exist for conversion of compounds (IX) and (X) to compounds of Formulas (I) and (II), respectively. Accordingly, preparation of (I) and (II) from carboxylic acids (IX) and (X) are well within the ambit of the skilled artisan.
  • Compositions and Methods of Administration
  • The compositions provided herein contain therapeutically effective amounts of one or more of the compounds provided herein that are useful in the prevention, treatment, or amelioration of one or more of the symptoms of diseases or disorders described herein and a vehicle. Vehicles suitable for administration of the compounds provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration.
  • In addition, the compounds may be formulated as the sole active ingredient in the composition or may be combined with other active ingredients.
  • The compositions contain one or more compounds provided herein. The compounds are, in some embodiments, formulated into suitable preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as topical administration, transdermal administration and oral inhalation via nebulizers, pressurized metered dose inhalers and dry powder inhalers. In some embodiments, the compounds described above are formulated into compositions using techniques and procedures well known in the art (see, e.g., Ansel Introduction to Pharmaceutical Dosage Forms, Seventh Edition (1999).
  • In the compositions, effective concentrations of one or more compounds or derivatives thereof is (are) mixed with a suitable vehicle. The compounds may be derivatized as the corresponding salts, esters, enol ethers or esters, acetals, ketals, orthoesters, hemiacetals, hemiketals, acids, bases, solvates, ion-pairs, hydrates or prodrugs prior to formulation, as described above. The concentrations of the compounds in the compositions are effective for delivery of an amount, upon administration that treats, leads to prevention, or amelioration of one or more of the symptoms of diseases or disorders described herein. In some embodiments, the compositions are formulated for single dosage administration. To formulate a composition, the weight fraction of a compound is dissolved, suspended, dispersed or otherwise mixed in a selected vehicle at an effective concentration such that the treated condition is relieved, prevented, or one or more symptoms are ameliorated.
  • The active compound is included in the vehicle in an amount sufficient to exert a therapeutically useful effect in the absence of undesirable side effects on the patient treated. The therapeutically effective concentration may be predicted empirically by testing the compounds in in vitro and in vivo systems well known to those of skill in the art and then extrapolated therefrom for dosages for humans. Human doses are then typically fine-tuned in clinical trials and titrated to response.
  • The concentration of active compound in the composition will depend on absorption, inactivation and excretion rates of the active compound, the physicochemical characteristics of the compound, the dosage schedule, and amount administered as well as other factors known to those of skill in the art. For example, the amount that is delivered is sufficient to ameliorate one or more of the symptoms of diseases or disorders as described herein.
  • In some embodiments, a therapeutically effective dosage should produce a serum concentration of active ingredient of from about 0.001 ng/ml to about 50-200 μg/ml. The compositions, in other embodiments, should provide a dosage of from about 0.0001 mg to about 70 mg of compound per kilogram of body weight per day. Dosage unit forms are prepared to provide from about 0.01 mg, 0.1 mg or 1 mg to about 500 mg, 1000 mg or 5000 mg, and in some embodiments from about 10 mg to about 500 mg of the active ingredient or a combination of essential ingredients per dosage unit form.
  • The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment is a function of the disease being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data or subsequent clinical testing. It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.
  • In instances in which the compounds exhibit insufficient solubility, methods for solubilizing compounds may be used such as use of liposomes, prodrugs, complexation/chelation, nanoparticles, or emulsions or tertiary templating. Such methods are known to those of skill in this art, and include, but are not limited to, using co-solvents, such as dimethylsulfoxide (DMSO), using surfactants or surface modifiers, such as TWEEN®, complexing agents such as cyclodextrin or dissolution by enhanced ionization (i.e. dissolving in aqueous sodium bicarbonate). Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective compositions.
  • Upon mixing or addition of the compound(s), the resulting mixture may be a solution, suspension, emulsion or the like. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected vehicle. The effective concentration is sufficient for ameliorating the symptoms of the disease, disorder or condition treated and may be empirically determined
  • The compositions are provided for administration to humans and animals in indication appropriate dosage forms, such as dry powder inhalers (DPIs), pressurized metered dose inhalers (pMDIs), nebulizers, tablets, capsules, pills, sublingual tapes/bioerodible strips, tablets or capsules, powders, granules, lozenges, lotions, salves, suppositories, fast melts, transdermal patches or other transdermal application devices/preparations, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the compounds or derivatives thereof. The therapeutically active compounds and derivatives thereof are, in some embodiments, formulated and administered in unit-dosage forms or multiple-dosage forms. Unit-dose forms as used herein refer to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required vehicle. Examples of unit-dose forms include ampoules and syringes and individually packaged tablets or capsules. Unit-dose forms may be administered in fractions or multiples thereof. A multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pints or gallons. Hence, multiple dose form is a multiple of unit-doses which are not segregated in packaging.
  • Liquid compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional adjuvants in a vehicle, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension, colloidal dispersion, emulsion or liposomal formulation. If desired, the composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 15th Edition, 1975 or later editions thereof.
  • Dosage forms or compositions containing active ingredient in the range of 0.005% to 100% with the balance made up from vehicle or carrier may be prepared. Methods for preparation of these compositions are known to those skilled in the art. The contemplated compositions may contain 0.001%-100% active ingredient, in one embodiment 0.1-95%, in another embodiment 0.4-10%.
  • In certain embodiments, the compositions are lactose-free compositions containing excipients that are well known in the art and are listed, for example, in the U.S. Pharmacopeia (USP) 25-NF20 (2002). In general, lactose-free compositions contain active ingredients, a binder/filler, and a lubricant in compatible amounts. Particular lactose-free dosage forms contain active ingredients, microcrystalline cellulose, pre-gelatinized starch, and magnesium stearate.
  • Further provided are anhydrous compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds. For example, the addition of water (e.g., 5%) is widely accepted as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, N.Y., 1995, pp. 379-80. In effect, water and heat accelerate the decomposition of some compounds. Thus, the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
  • Anhydrous compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • An anhydrous composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are generally packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
  • Oral dosage forms are either solid, gel or liquid. The solid dosage forms are tablets, capsules, granules, and bulk powders. Types of oral tablets include compressed, chewable lozenges and tablets which may be enteric-coated, sugar-coated or film-coated. Capsules may be hard or soft gelatin capsules, while granules and powders may be provided in non-effervescent or effervescent form with the combination of other ingredients known to those skilled in the art.
  • In certain embodiments, the formulations are solid dosage forms such as for example, capsules or tablets. The tablets, pills, capsules, troches and the like can contain one or more of the following ingredients, or compounds of a similar nature: a binder; a lubricant; a diluent; a glidant; a disintegrating agent; a coloring agent; a sweetening agent; a flavoring agent; a wetting agent; an enteric coating; a film coating agent and modified release agent. Examples of binders include microcrystalline cellulose, methyl paraben, polyalkyleneoxides, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, molasses, polyvinylpyrrolidine, povidone, crospovidones, sucrose and starch and starch derivatives. Lubricants include talc, starch, magnesium/calcium stearate, lycopodium and stearic acid. Diluents include, for example, lactose, sucrose, trehalose, lysine, leucine, lecithin, starch, kaolin, salt, mannitol and dicalcium phosphate. Glidants include, but are not limited to, colloidal silicon dioxide. Disintegrating agents include crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose. Coloring agents include, for example, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate and advanced coloring or anti-forgery color/opalescent additives known to those skilled in the art. Sweetening agents include sucrose, lactose, mannitol and artificial sweetening agents such as saccharin, and any number of spray dried flavors. Flavoring agents include natural flavors extracted from plants such as fruits and synthetic blends of compounds which produce a pleasant sensation or mask unpleasant taste, such as, but not limited to peppermint and methyl salicylate. Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene laural ether. Enteric-coatings include fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates. Film coatings include hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate. Modified release agents include polymers such as the Eudragit® series and cellulose esters.
  • The compound, or derivative thereof, can be provided in a composition that protects it from the acidic environment of the stomach. For example, the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine. The composition may also be formulated in combination with an antacid or other such ingredient.
  • When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents. The compounds can also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like. A syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.
  • The active materials can also be mixed with other active materials which do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics. The active ingredient is a compound or derivative thereof as described herein. Higher concentrations, up to about 98% by weight of the active ingredient may be included.
  • In all embodiments, tablets and capsules formulations may be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient. Thus, for example, they may be coated with a conventional enterically digestible coating, such as phenylsalicylate, waxes and cellulose acetate phthalate.
  • Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules. Aqueous solutions include, for example, elixirs and syrups. Emulsions are either oil-in-water or water-in-oil.
  • Elixirs are clear, sweetened, hydroalcoholic preparations. Vehicles used in elixirs include solvents. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may contain a preservative. An emulsion is a two-phase system in which one liquid is dispersed in the form of small globules throughout another liquid. Carriers used in emulsions are non-aqueous liquids, emulsifying agents and preservatives. Suspensions use suspending agents and preservatives. Acceptable substances used in non-effervescent granules, to be reconstituted into a liquid oral dosage form, include diluents, sweeteners and wetting agents. Acceptable substances used in effervescent granules, to be reconstituted into a liquid oral dosage form, include organic acids and a source of carbon dioxide. Coloring and flavoring agents are used in all of the above dosage forms.
  • Solvents include glycerin, sorbitol, ethyl alcohol and syrup. Examples of preservatives include glycerin, methyl and propylparaben, benzoic acid, sodium benzoate and alcohol. Examples of non-aqueous liquids utilized in emulsions include mineral oil and cottonseed oil. Examples of emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate. Suspending agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum and acacia. Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as saccharin. Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether. Organic acids include citric and tartaric acid. Sources of carbon dioxide include sodium bicarbonate and sodium carbonate. Coloring agents include any of the approved certified water soluble FD and C dyes, and mixtures thereof. Flavoring agents include natural flavors extracted from plants such fruits, and synthetic blends of compounds which produce a pleasant taste sensation.
  • For a solid dosage form, the solution or suspension, in for example, propylene carbonate, vegetable oils or triglycerides, is in some embodiments encapsulated in a gelatin capsule. Such solutions, and the preparation and encapsulation thereof, are disclosed in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545. For a liquid dosage form, the solution, e.g., for example, in a polyethylene glycol, may be diluted with a sufficient quantity of a liquid vehicle, e.g., water, to be easily measured for administration.
  • Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g., propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells. Other useful formulations include those set forth in U.S. Pat. Nos. RE28,819 and 4,358,603. Briefly, such formulations include, but are not limited to, those containing a compound provided herein, a dialkylated mono- or polyalkylene glycol, including, but not limited to, 1,2-dimethoxyethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether wherein 350, 550 and 750 refer to the approximate average molecular weight of the polyethylene glycol, and one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, thiodipropionic acid and its esters, and dithiocarbamates.
  • Other formulations include, but are not limited to, aqueous alcoholic solutions including a acetal. Alcohols used in these formulations are any water-miscible solvents having one or more hydroxyl groups, including, but not limited to, propylene glycol and ethanol. Acetals include, but are not limited to, di(lower alkyl) acetals of lower alkyl aldehydes such as acetaldehyde diethyl acetal.
  • Parenteral administration, in some embodiments characterized by injection, either subcutaneously, intramuscularly or intravenously is also contemplated herein. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. The injectables, solutions and emulsions also contain one or more excipients. Suitable excipients are, for example, water, saline, dextrose, glycerol or ethanol. In addition, if desired, the compositions to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.
  • Implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained (see, e.g., U.S. Pat. No. 3,710,795) is also contemplated herein. Briefly, a compound provided herein is dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer, that is insoluble in body fluids. The compound diffuses through the outer polymeric membrane in a release rate controlling step. The percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject.
  • Parenteral administration of the compositions includes intravenous, subcutaneous and intramuscular administrations. Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as lyophilized powders, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile emulsions. The solutions may be either aqueous or nonaqueous.
  • If administered intravenously, suitable carriers include physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.
  • Vehicles used in parenteral preparations include aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other substances.
  • Examples of aqueous vehicles include Sodium Chloride Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection, Dextrose and Lactated Ringers Injection. Nonaqueous parenteral vehicles include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil. Antimicrobial agents in bacteriostatic or fungistatic concentrations must be added to parenteral preparations packaged in multiple-dose containers which include phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride. Isotonic agents include sodium chloride and dextrose. Buffers include phosphate and citrate. Antioxidants include sodium bisulfate. Local anesthetics include procaine hydrochloride. Suspending and dispersing agents include sodium carboxymethylcelluose, hydroxypropyl methylcellulose and polyvinylpyrrolidone. Emulsifying agents include Polysorbate 80 (Tween® 80). A sequestering or chelating agent of metal ions includes EDTA. Carriers also include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles; and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.
  • The concentration of compound is adjusted so that an injection provides an effective amount to produce the desired pharmacological effect. The exact dose depends on the age, weight, body surface area and condition of the patient or animal as is known in the art.
  • The unit-dose parenteral preparations are packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration must be sterile, as is known and practiced in the art.
  • Illustratively, intravenous or intraarterial infusion of a sterile aqueous solution containing an active compound is an effective mode of administration. Another embodiment is a sterile aqueous or oily solution or suspension containing an active material injected as necessary to produce the desired pharmacological effect.
  • Injectables are designed for local and systemic administration. In some embodiments, a therapeutically effective dosage is formulated to contain a concentration of at least about 0.01% w/w up to about 90% w/w or more, in certain embodiments more than 0.1% w/w of the active compound to the treated tissue(s).
  • The compound may be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the condition and may be empirically determined.
  • Active ingredients provided herein can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,639,480; 5,733,566; 5,739,108; 5,891,474; 5,922,356; 5,972,891; 5,980,945; 5,993,855; 6,045,830; 6,087,324; 6,113,943; 6,197,350; 6,248,363; 6,264,970; 6,267,981; 6,376,461; 6,419,961; 6,589,548; 6,613,358; 6,699,500 and 6,740,634. Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients provided herein.
  • All controlled-release products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts. Ideally, the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time. Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance. In addition, controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Most controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release of other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body. Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • In certain embodiments, the agent may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration. In some embodiments, a pump may be used (see, Sefton, CRC Crit. Ref Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)). In other embodiments, polymeric materials can be used. In other embodiments, a controlled release system can be placed in proximity of the therapeutic target, i.e., thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical Applications of Controlled Release, vol. 2, pp. 115-138 (1984)). In some embodiments, a controlled release device is introduced into a subject in proximity of the site of inappropriate immune activation or a tumor. Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)). The active ingredient can be dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross-linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer, that is insoluble in body fluids. The active ingredient then diffuses through the outer polymeric membrane in a release rate controlling step. The percentage of active ingredient contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the needs of the subject.
  • Of interest herein are also lyophilized powders, which can be reconstituted for administration as solutions, emulsions and other mixtures. They may also be reconstituted and formulated as solids or gels.
  • The sterile, lyophilized powder is prepared by dissolving a compound provided herein, or a derivative thereof, in a suitable solvent. The solvent may contain an excipient which improves the stability or other pharmacological component of the powder or reconstituted solution, prepared from the powder. Excipients that may be used include, but are not limited to, an antioxidant, a buffer and a bulking agent. In some embodiments, the excipient is selected from dextrose, sorbital, fructose, corn syrup, xylitol, glycerin, glucose, sucrose and other suitable agent. The solvent may contain a buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, at about neutral pH. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation. In some embodiments, the resulting solution will be apportioned into vials for lyophilization. Each vial will contain a single dosage or multiple dosages of the compound. The lyophilized powder can be stored under appropriate conditions, such as at about 4° C. to room temperature.
  • Reconstitution of this lyophilized powder with water for injection provides a formulation for use in parenteral administration. For reconstitution, the lyophilized powder is added to sterile water or other suitable carrier. The precise amount depends upon the selected compound. Such amount can be empirically determined
  • Topical mixtures are prepared as described for the local and systemic administration. The resulting mixture may be a solution, suspension, emulsions or the like and are formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical administration.
  • The compounds or derivatives thereof may be formulated as aerosols for topical application, such as by inhalation (see, e.g., U.S. Pat. Nos. 4,044,126, 4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment of inflammatory diseases, particularly asthma). These formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microtine powder for insufflation, alone or in combination with an inert carrier such as lactose. In such a case, the particles of the formulation will, in some embodiments, have mass median geometric diameters of less than 5 microns, in other embodiments less than 10 microns.
  • Oral inhalation formulations of the compounds or derivatives suitable for inhalation include metered dose inhalers, dry powder inhalers and liquid preparations for administration from a nebulizer or metered dose liquid dispensing system. For both metered dose inhalers and dry powder inhalers, a crystalline form of the compounds or derivatives is the preferred physical form of the drug to confer longer product stability.
  • In addition to particle size reduction methods known to those skilled in the art, crystalline particles of the compounds or derivatives can be generated using supercritical fluid processing which offers significant advantages in the production of such particles for inhalation delivery by producing respirable particles of the desired size in a single step. (e.g., International Publication No. WO2005/025506). A controlled particle size for the microcrystals can be selected to ensure that a significant fraction of the compounds or derivatives is deposited in the lung. In some embodiments, these particles have a mass median aerodynamic diameter of about 0.1 to about 10 microns, in other embodiments, about 1 to about 5 microns and still other embodiments, about 1.2 to about 3 microns.
  • Inert and non-flammable HFA propellants are selected from HFA 134a (1,1,1,2-tetrafluoroethane) and HFA 227e (1,1,1,2,3,3,3-heptafluoropropane) and provided either alone or as a ratio to match the density of crystal particles of the compounds or derivatives. A ratio is also selected to ensure that the product suspension avoids detrimental sedimentation or cream (which can precipitate irreversible agglomeration) and instead promote a loosely flocculated system, which is easily dispersed when shaken. Loosely fluctuated systems are well regarded to provide optimal stability for pMDI canisters. As a result of the formulation's properties, the formulation contained no ethanol and no surfactants/stabilizing agents.
  • The formulation of the compounds or derivatives can be administered to patients using TEMPO® (MAP Pharmaceuticals, Inc., Mountain View, Calif.), a novel breath activated metered dose inhaler. TEMPO® overcomes the variability associated with standard pressurized metered dose inhalers (pMDI), and achieves consistent delivery of drug to the lung periphery where it can be systemically absorbed. To do so, TEMPO® incorporates four novel features: 1) breath synchronous trigger—can be adjusted for different drugs and target populations to deliver the drug at a specific part of the inspiratory cycle, 2) plume control—an impinging jet to slow down the aerosol plume within the actuator, 3) vortexing chamber—consisting of porous wall, which provides an air cushion to keep the slowed aerosol plume suspended and air inlets on the back wall which drive the slowed aerosol plume into a vortex pattern, maintaining the aerosol in suspension and allowing the particle size to reduce as the HFA propellant evaporates, and 4) dose counter—will determine the doses remaining and prevent more than the intended maximum dose to be administered from any one canister.
  • The compounds may be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application. Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies. Nasal solutions of the active compound alone or in combination with other excipients can also be administered.
  • For nasal administration, the preparation may contain an esterified phosphonate compound dissolved or suspended in a liquid carrier, in particular, an aqueous carrier, for aerosol application. The carrier may contain solubilizing or suspending agents such as propylene glycol, surfactants, absorption enhancers such as lecithin or cyclodextrin, or preservatives.
  • Solutions, particularly those intended for ophthalmic use, may be formulated as 0.01%-10% isotonic solutions, pH about 5-7.4, with appropriate salts.
  • Other routes of administration, such as transdermal patches, including iontophoretic and electrophoretic devices, and rectal administration, are also contemplated herein.
  • Transdermal patches, including iotophoretic and electrophoretic devices, are well known to those of skill in the art. For example, such patches are disclosed in U.S. Pat. Nos. 6,267,983, 6,261,595, 6,256,533, 6,167,301, 6,024,975, 6,010715, 5,985,317, 5,983,134, 5,948,433 and 5,860,957.
  • For example, dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect. Rectal suppositories are used herein mean solid bodies for insertion into the rectum which melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients. Substances utilized in rectal suppositories are bases or vehicles and agents to raise the melting point. Examples of bases include cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol) and appropriate mixtures of mono-, di- and triglycerides of fatty acids. Combinations of the various bases may be used. Agents to raise the melting point of suppositories include spermaceti and wax. Rectal suppositories may be prepared either by the compressed method or by molding. The weight of a rectal suppository, in one embodiment, is about 2 to 3 gm. Tablets and capsules for rectal administration are manufactured using the same substance and by the same methods as for formulations for oral administration.
  • The compounds provided herein, or derivatives thereof, may also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated. Many such targeting methods are well known to those of skill in the art. All such targeting methods are contemplated herein for use in the instant compositions. For non-limiting examples of targeting methods, see, e.g., U.S. Pat. Nos. 6,316,652, 6,274,552, 6,271,359, 6,253,872, 6,139,865, 6,131,570, 6,120,751, 6,071,495, 6,060,082, 6,048,736, 6,039,975, 6,004,534, 5,985,307, 5,972,366, 5,900,252, 5,840,674, 5,759,542 and 5,709,874.
  • In some embodiments, liposomal suspensions, including tissue-targeted liposomes, such as tumor-targeted liposomes, may also be suitable as carriers. These may be prepared according to methods known to those skilled in the art. For example, liposome formulations may be prepared as described in U.S. Pat. No. 4,522,811. Briefly, liposomes such as multilamellar vesicles (MLV's) may be formed by drying down phosphatidyl choline and phosphatidyl serine (7:3 molar ratio) on the inside of a flask. A solution of a compound provided herein in phosphate buffered saline lacking divalent cations (PBS) is added and the flask shaken until the lipid film is dispersed. The resulting vesicles are washed to remove unencapsulated compound, pelleted by centrifugation, and then resuspended in PBS.
  • The compounds or derivatives may be packaged as articles of manufacture containing packaging material, a compound or derivative thereof provided herein, which is effective for treatment, prevention or amelioration of one or more symptoms of the diseases or disorders, supra, within the packaging material, and a label that indicates that the compound or composition or derivative thereof, is used for the treatment, prevention or amelioration of one or more symptoms of the diseases or disorders, supra.
  • The articles of manufacture provided herein contain packaging materials. Packaging materials for use in packaging products are well known to those of skill in the art. See, e.g., U.S. Pat. Nos. 5,323,907, 5,052,558 and 5,033,252. Examples of packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment. A wide array of formulations of the compounds and compositions provided herein are contemplated as are a variety of treatments for any disease or disorder described herein.
  • Dosages
  • In human therapeutics, the physician will determine the dosage regimen that is most appropriate according to a preventive or curative treatment and according to the age, weight, stage of the disease and other factors specific to the subject to be treated. The compositions, in other embodiments, should provide a dosage of from about 0.0001 mg to about 70 mg of compound per kilogram of body weight per day. Dosage unit forms are prepared to provide from about 0.01 mg, 0.1 mg or 1 mg to about 500 mg, 1000 mg or 5000 mg, and in some embodiments from about 10 mg to about 500 mg of the active ingredient or a combination of essential ingredients per dosage unit form. The amount of active ingredient in the formulations provided herein, which will be effective in the prevention or treatment of a disorder or one or more symptoms thereof, will vary with the nature and severity of the disease or condition, and the route by which the active ingredient is administered. The frequency and dosage will also vary according to factors specific for each subject depending on the specific therapy (e.g., therapeutic or prophylactic agents) administered, the severity of the disorder, disease, or condition, the route of administration, as well as age, body, weight, response, and the past medical history of the subject.
  • Exemplary doses of a formulation include milligram or microgram amounts of the active compound per kilogram of subject (e.g., from about 1 micrograms per kilogram to about 50 milligrams per kilogram, from about 10 micrograms per kilogram to about 30 milligrams per kilogram, from about 100 micrograms per kilogram to about 10 milligrams per kilogram, or from about 100 microgram per kilogram to about 5 milligrams per kilogram).
  • It may be necessary to use dosages of the active ingredient outside the ranges disclosed herein in some cases, as will be apparent to those of ordinary skill in the art. Furthermore, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with subject response.
  • Different therapeutically effective amounts may be applicable for different diseases and conditions, as will be readily known by those of ordinary skill in the art. Similarly, amounts sufficient to prevent, manage, treat or ameliorate such disorders, but insufficient to cause, or sufficient to reduce, adverse effects associated with the composition provided herein are also encompassed by the above described dosage amounts and dose frequency schedules. Further, when a subject is administered multiple dosages of a composition provided herein, not all of the dosages need be the same. For example, the dosage administered to the subject may be increased to improve the prophylactic or therapeutic effect of the composition or it may be decreased to reduce one or more side effects that a particular subject is experiencing.
  • In certain embodiments, administration of the same formulation provided herein may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • Methods of Use of the Compounds and Compositions
  • Methods of treating, preventing, or ameliorating one or more symptoms of diseases including, for example, central nervous system disorders, migraine, and Parkinson's disease are also provided herein. In practicing the methods, therapeutically effective amounts of the compounds or compositions, described herein, supra, are administered.
  • Also provided are methods and compositions for decreased 5-HT2 receptor agonist activity. In some embodiment, the 5-HT2A and 5-HT2B activity agonist activity is lower than the 5-HT2A and 5-HT2B reported for cabergoline. Also provided are methods for antagonizing receptors including 5-HT2A and 5-HT2B receptors using the compounds and compositions, described herein. In practicing the methods, therapeutically effective amounts of the compounds or compositions, described herein, supra, are administered.
  • In some embodiments, it is desirable to select compounds that are useful for the treatment of one or more symptoms of Parkinson's disease. An ideal compound for such treatment should have selective agonist activities for the dopaminergic D2 receptor. In some cases, it would be preferable for the compound to have a stronger affinity (and agonist activity) for the D2 receptor compared to D1 and D4 receptors. Additionally, in some embodiments, it may be advantageous for a compound to have weak to moderate 5-HT1B and 5-HT1D receptor agonist activities. In other embodiments, it may be advantageous for a compound to have 5-HT2 receptors antagonism activities. As an illustrative example (and described in detail in the Examples below), the cabergoline-derivative compound, hereinafter referred to as Compound 1,
  • Figure US20150202198A1-20150723-C00011
  • exhibited weak serotonin 5-HT2A and 5-HT2B receptor agonist activity (as compared to cabergoline). Additionally, Compound 1 is selective for the dopaminergic receptor D2L, compared to D1 and D4 receptors. Compound 1 also exhibited weak agonist activity on serotonin receptor 5-HT1D. Compound 1 exhibited properties that are more desirable than cabergoline in that 5-HT2A agonism is associated with hallucination, which is often a side effect of some anti-Parkinson's agents (see for example, Egan et al., Psychoparmacology (1998) 136:409-414). Agonist activity of the 5-HT2B receptor has also been shown to be associated with cardiac and non-cardiac fibrosis. The lower 5-HT2B agonist activity seen with Compound 1 (compared to cabergoline) may translate into minimal or a reduction in the undesired fibrosis side-effects seen with cabergoline usage. Without being bound to any particular theory, it is hypothesized that the CF3 modification in Compound 1 (compared to cabergoline) is responsible for the desirable effects of this molecule.
  • Depending on the dosing or treatment regimen, in some embodiments, it may be advantageous for the compound to have a relatively short half-life. In some embodiments, the half-life of the compound is less than 5 hours. In other embodiments, the half-life of the compound is between 1 hour and 5 hours. In still other embodiments, the half-life of the compound is less than 8 hours. Alternatively, it may be advantageous for the compound to have a moderate to long half-life. In some embodiments, the half-life of the compound is greater than 10 hours. In other embodiments, the half-life of the compound is between 12 and 24 hours.
  • Combination Therapy
  • The compounds and compositions disclosed herein may also be used in combination with one or more other active ingredients. In certain embodiments, the compounds may be administered in combination, or sequentially, with another therapeutic agent. Such other therapeutic agents include, for example, those known for treatment, prevention, or amelioration of one or more symptoms associated with migraine or Parkinson's disease. As an illustrative example, compounds such as Compound 1 may be administered to a patient with Parkinson's disease as an adjunct therapy in addition to monotherapies, such as L-dihydroxyphenylacetyl acid (L-DOPA). Alternatively, compounds such as Compound 1 may be administered as a monotherapy.
  • It should be understood that any suitable combination of the compounds and compositions provided herein with one or more of the above therapeutic agents and optionally one or more further pharmacologically active substances are considered to be within the scope of the present disclosure. In some embodiments, the compounds and compositions provided herein are administered prior to or subsequent to the one or more additional active ingredients.
  • It should also be understood that any suitable combination of the compounds and compositions provided herein may be used with other agents to agonize and or antagonize the receptors mentioned above.
  • Finally, it should be noted that there are alternative ways of implementing the present invention. Accordingly, the present embodiments are to be considered as illustrative and not restrictive, and the invention is not to be limited to the details given herein, but may be modified within the scope and equivalents of the appended claims.
  • All publications and patents cited herein are incorporated by reference in their entirety.
  • The following examples are provided for illustrative purposes only and are not intended to limit the scope of the invention.
  • EXAMPLES Example 1 Preparation of Compound 1
  • Compound 1 was synthesized using the following synthesis route:
  • Figure US20150202198A1-20150723-C00012
  • To a solution of Togni's reagent (63 mg, 0.199 mmol) and copper (I) acetate (2 mg, 0.016 mmol) in anhydrous methanol (3 mL) under an argon atmosphere was added cabergoline (75 mg, 0.166 mmol). The reaction mixture was heated at 40° C. for 4.5 hours and then allowed to cool to ambient temperature. The cooled mixture was then diluted with excess dichloromethane and washed with saturated aqueous NaHCO3. The dichloromethane layer was dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the crude product. The crude product was partially purified by flash column chromatography (SiO2, 90:10 to 85:15 v/v dichloromethane/methanol). The product was then subjected to further purification by semi-preparative HPLC to afford Compound 1 as a light yellow solid. 1H NMR results for Compound 1 was as follows: 1H NMR (CDCl3, 400 MHz) δ 9.45 (br s, 1H), 8.23 (s, 1H), 7.28 (d, J=8.0 Hz, 1H), 7.20 (d, J=8.0 Hz, 1H), 6.95 (d, J=6.8 Hz, 1H), 5.90-6.00 (m, 1H), 5.26 (dd, J=17.2, 1.2 Hz, 1H), 5.20 (d, J=10.0 Hz, 1H), 3.72-3.92 (m, 2H), 3.53-3.60 (m, 2H), 3.27-3.48 (m, 4H), 3.15-3.20 (m, 1H), 2.88-2.97 (m, 1H), 2.68-2.83 (m, 2H), 2.53-2.63 (m, 2H), 2.30-2.42 (m, 2H), 2,24 (s, 6H), 1.82-1.91 (m, 2H), 1.70-1.80 (m, 1H), 1.19 (t, J=7.2 Hz, 3H); 19F NMR (CDCl3, 282 MHz) δ −59.2; APCI MS m/z 520 [M+H]+.
  • Example 2 Receptor Agonist/Antagonist Action of Compound 1
  • Compound 1 was synthesized as described above and assayed for receptor activity against a panel of dopaminergic and serotoninergic receptors. Recombinant human serotonin receptors 5-HT2A, and 5-HT2B were expressed in CHO-K1 cells and activity (receptor agonist/antagonist activity) was assayed using an Aequorin assay (coexpression of mitochondrial apoaequorin). Compound 1 was used at a concentration of 0.010 nM to 20,000 nM for agonist activity and 0.005 nM to 10,000 nM for antagonist activity. Results showed that Compound 1 displayed much less activity on 5-HT2A and 5-HT2B receptors as compared to cabergoline. On 5-HT2A receptors, Compound 1 had an EC50 of 946 nM, compared to the reported EC50 of 7.8 nM of cabergoline. On 5-HT2B receptors, Compound 1 had an EC50 of 6049 nM, compared to the reported EC50 of 2.6 nM of cabergoline (Newman-Tancredi, et al., J Pharmacology & Experimental Therapeutics (2002) 303(2): 815-822). The results from the receptor assays on human serotonin receptors indicate that Compound 1 may have a decreased or minimized occurrence of the undesired cardiac and non-cardiac fibrosis side-effects that are associated with cabergoline.
  • Recombinant human dopaminergic receptor D1, D2L, and D4 and serotonin receptor 5-HT1B and 5-HT1D activity was assayed using a GTPγS functional assay with Compound 1 at concentrations between 0.01 nM and 10,000 nM. Results showed that Compound 1 exhibited strong agonist activities with the D2L receptor (EC50 of 12.6 nM). Additionally, Compound 1 exhibited an 18-fold D2L to D4 receptor selectivity (when comparing EC50). Compared to reported results for cabergoline, Compound 1 has a lower fold selectivity for D2L than D4, cabergoline has approximately a 200-fold selectivity for the D2L to D4 receptor selectivity. However, Compound 1 also exhibited a lower Emax (26%) as compared to reported values for cabergoline (Emax of 49%) (Newman-Tancredi, et al., J Pharmacology & Experimental Therapeutics (2002) 303(2): 805-814). Compound 1 was also highly selective for D2L compared to D1 receptors, with results showing approximately 110-fold selectivity for the D2L receptor over the D1 receptor. For the serotonin receptors, 5-HT1B and 5-HT1D, Compound 1 was not active on 5-HT1B receptors, whereas data have shown that cabergoline is active and has an EC50 of 2238 nM (see for example, Newman-Tancredi, et al., J Pharmacology & Experimental Therapeutics (2002) 303(2): 815-822. Compound 1 was an agonist on 5-HT1D receptors with an EC50 of 263 nM, which shows less affinity than cabergoline (EC50 of 16 nM; Newman-Tancredi, et al., J Pharmacology & Experimental Therapeutics (2002) 303(2): 815-822). In addition, Compound 1 had a reduced Emax from 68% to 31% compared to cabergoline. Taken together, these data is consistent with the conclusion that Compound 1 would be a desirable anti-Parkinson's disease agent.

Claims (12)

What is claimed is:
1. A method of treating and/or preventing central nervous system disorders in a subject comprising administering to the subject in need thereof a therapeutically effective amount of the compound having the structure:
Figure US20150202198A1-20150723-C00013
wherein R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R5 is hydrogen or (C1-C3) alkyl, and R11 is (C1-C3) alkyl substituted with one or more fluorine atoms.
2. The method of claim 1, wherein R1 is hydrogen, —CH3 or —CF3, R5 is hydrogen, methyl or allyl, and R11 is —CF3.
3. The method of claim 1, wherein R1 is —CH3 or —CF3, R5 is methyl or allyl, and R11 is —CF3.
4. The method of claim 1, wherein R1 is —CH3 or —CF3, R5 is methyl or allyl, and R11 is —CF3.
5. A method of treating and/or preventing migraine in a subject comprising administering to the subject in need thereof a therapeutically effective amount of the compound having the structure:
Figure US20150202198A1-20150723-C00014
wherein R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R5 is hydrogen or (C1-C3) alkyl, and R11 is (C1-C3) alkyl substituted with one or more fluorine atoms.
6. The method of claim 5, wherein R1 is hydrogen, —CH3 or —CF3, R5 is hydrogen, methyl or allyl, and R11 is —CF3.
7. The method of claim 5, wherein R1 is —CH3 or —CF3, R5 is methyl or allyl, and R11 is —CF3.
8. The method of claim 5, wherein R1 is —CH3 or —CF3, R5 is methyl or allyl, and R11 is —CF3.
9. A method of treating and/or preventing Parkinson's disease in a subject comprising administering to the subject in need thereof a therapeutically effective amount of the compound having the structure:
Figure US20150202198A1-20150723-C00015
wherein R1 is hydrogen, (C1-C4) alkyl or (C1-C4) alkyl substituted with one or more fluorine atoms, R5 is hydrogen or (C1-C3) alkyl, and R11 is (C1-C3) alkyl substituted with one or more fluorine atoms.
10. The method of claim 9, wherein R1 is hydrogen, —CH3 or —CF3, R5 is hydrogen, methyl or allyl, and R11 is —CF3.
11. The method of claim 9, wherein R1 is —CH3 or —CF3, R5 is methyl or allyl, and R11 is —CF3.
12. The method of claim 9, wherein R1 is —CH3 or —CF3, R5 is methyl or allyl, and R11 is —CF3.
US14/670,953 2012-06-22 2015-03-27 Novel Cabergoline Derivatives Abandoned US20150202198A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/670,953 US20150202198A1 (en) 2012-06-22 2015-03-27 Novel Cabergoline Derivatives

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13/531,432 US9012640B2 (en) 2012-06-22 2012-06-22 Cabergoline derivatives
US14/670,953 US20150202198A1 (en) 2012-06-22 2015-03-27 Novel Cabergoline Derivatives

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/531,432 Division US9012640B2 (en) 2012-06-22 2012-06-22 Cabergoline derivatives

Publications (1)

Publication Number Publication Date
US20150202198A1 true US20150202198A1 (en) 2015-07-23

Family

ID=49774937

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/531,432 Active 2033-08-08 US9012640B2 (en) 2012-06-22 2012-06-22 Cabergoline derivatives
US14/670,953 Abandoned US20150202198A1 (en) 2012-06-22 2015-03-27 Novel Cabergoline Derivatives

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/531,432 Active 2033-08-08 US9012640B2 (en) 2012-06-22 2012-06-22 Cabergoline derivatives

Country Status (1)

Country Link
US (2) US9012640B2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11133224B2 (en) * 2019-09-27 2021-09-28 Taiwan Semiconductor Manufacturing Co., Ltd. Semiconductor structure and method for forming the same

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2013015373A (en) * 2011-06-23 2014-02-11 Map Pharmaceuticals Inc Novel fluoroergoline analogs.
WO2013095707A1 (en) * 2011-12-19 2013-06-27 Map Pharmaceuticals, Inc. Novel iso-ergoline derivatives
MX2017009405A (en) 2015-01-20 2018-01-18 Xoc Pharmaceuticals Inc Ergoline compounds and uses thereof.
BR112017015510A2 (en) 2015-01-20 2018-01-30 Xoc Pharmaceuticals Inc compound of formula (i), method of treatment and / or prevention, d2 receptor agonizing method in one individual, d3 receptor antagonizing method in one individual, 5-ht1d receptor agonizing method in one individual, 5-ht1a receptor agonization in one individual, selective 5-ht1d receptor agonizing method instead of 5-ht1b receptor in one individual, 5-ht2c re-receptor selective agonizing method instead of 5-ht2a or 5 receptor -ht2b in one individual, method of 5-ht2c receptor agonization in one individual, method of providing functional antagonist activity at 5-ht2b receptor or 5-ht7 receptor, and, method of providing functional antagonist activity at adrenergic receptors in one individual
MX2019014272A (en) 2017-06-01 2020-12-11 Xoc Pharmaceuticals Inc Ergoline derivatives for use in medicine.

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040198753A1 (en) * 2002-01-28 2004-10-07 Hiroshi Kase Methods of treating patients suffering from movement disorders

Family Cites Families (113)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3190884A (en) 1965-06-22 Chz-chz-cn
CH344731A (en) 1956-05-18 1960-02-29 Sandoz Ag Process for the production of new derivatives of the lysergic acid series alkylated on the indole nitrogen
US2886568A (en) 1956-10-16 1959-05-12 Union Carbide Corp Preparation of quinolines
GB1011112A (en) 1961-05-10 1965-11-24 Sandoz Ag Improvements in or relating to ergot alkaloids
FR1583797A (en) 1967-04-12 1969-12-05
CH520680A (en) 1967-08-02 1972-03-31 Sandoz Ag Ergot derivs andrenolytics
CH508628A (en) 1968-06-25 1971-06-15 Sandoz Ag Process for the preparation of new heterocyclic compounds
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
GB1298277A (en) 1969-04-18 1972-11-29 Sandoz Ltd Preparation of ergot peptide alkaloids
CH517099A (en) 1969-06-20 1971-12-31 Sandoz Ag Process for the production of new ergot peptide alkaloids
US3710795A (en) 1970-09-29 1973-01-16 Alza Corp Drug-delivery device with stretched, rate-controlling membrane
US3814765A (en) 1970-11-12 1974-06-04 Farmaceutici Italia 8,9-didehydro-10-alkoxy-ergolenes and process of preparation thereof
US4165376A (en) 1971-01-29 1979-08-21 Lake Shore Roentgenology, Ltd. Treatment of the acute after-effects resulting from alcohol ingestion
US4044126A (en) 1972-04-20 1977-08-23 Allen & Hanburys Limited Steroidal aerosol compositions and process for the preparation thereof
GB1429184A (en) 1972-04-20 1976-03-24 Allen & Hanburys Ltd Physically anti-inflammatory steroids for use in aerosols
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
USRE28819E (en) 1972-12-08 1976-05-18 Syntex (U.S.A.) Inc. Dialkylated glycol compositions and medicament preparations containing same
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
HU169073B (en) 1974-05-28 1976-09-28
CH610330A5 (en) 1974-09-27 1979-04-12 Sandoz Ag Process for the preparation of novel ergopeptins
US3922347A (en) 1974-12-19 1975-11-25 Lilly Co Eli Method of inhibiting prolactin secretion with 8-acylaminoergolenes
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
GB1485738A (en) 1976-08-02 1977-09-14 Farmaceutici Italia Ergot alkaloids
US4124712A (en) 1976-09-06 1978-11-07 Sandoz Ltd. Ergot peptide alkaloid derivatives
GB1584464A (en) 1977-04-19 1981-02-11 Farmaceutici Italia Ergot alkaloids
US4410545A (en) 1981-02-13 1983-10-18 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4328245A (en) 1981-02-13 1982-05-04 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4358603A (en) 1981-04-16 1982-11-09 Syntex (U.S.A.) Inc. Acetal stabilized prostaglandin compositions
FI76085C (en) 1981-07-21 1988-09-09 Erba Farmitalia Process for the preparation of ergoline derivatives
CH649300A5 (en) 1981-08-07 1985-05-15 Sandoz Ag ERGOPEPTIN DERIVATIVES, THEIR PRODUCTION AND USE.
US4409239A (en) 1982-01-21 1983-10-11 Syntex (U.S.A.) Inc. Propylene glycol diester solutions of PGE-type compounds
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
HU196714B (en) 1984-10-04 1989-01-30 Monsanto Co Process for producing non-aqueous composition comprising somatotropin
IE58110B1 (en) 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
EG18586A (en) 1987-06-15 1993-08-30 Lilly Co Eli Cycloalkylamides of(beta,beta)-1-alkyl-6-(substituted)ergolines
US5033252A (en) 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
US5052558A (en) 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
US5668155A (en) 1988-05-10 1997-09-16 The General Hospital Corporation Administration of pirenzepine, methyl scopolamine and other muscarinic receptor antagonists for treatment of lipid metabolism disorders
DK338789A (en) 1988-07-15 1990-01-16 Schering Ag 2-SUBSTITUTED ERGOLINYLURINE DERIVATIVES AND PROCEDURES FOR THE PRODUCTION THEREOF, THEIR USE AS MEDICINES AND INTERMEDIATES FOR THE PRODUCTION THEREOF
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
IT1229203B (en) 1989-03-22 1991-07-25 Bioresearch Spa USE OF 5 METHYLTHETRAHYDROPHOLIC ACID, 5 FORMYLTHETRAHYDROPHOLIC ACID AND THEIR PHARMACEUTICALLY ACCEPTABLE SALTS FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS IN THE FORM OF CONTROLLED RELEASE ACTIVE IN THE THERAPY OF MENTAL AND ORGANIC DISORDERS.
PH30995A (en) 1989-07-07 1997-12-23 Novartis Inc Sustained release formulations of water soluble peptides.
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5585112A (en) 1989-12-22 1996-12-17 Imarx Pharmaceutical Corp. Method of preparing gas and gaseous precursor-filled microspheres
IT1246382B (en) 1990-04-17 1994-11-18 Eurand Int METHOD FOR THE TARGETED AND CONTROLLED DELIVERY OF DRUGS IN THE INTESTINE AND PARTICULARLY IN THE COLON
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5543390A (en) 1990-11-01 1996-08-06 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
DE4123587A1 (en) 1991-07-12 1993-01-14 Schering Ag 2,14-DISUBSTITUTED ERGOLINE, THEIR PRODUCTION AND USE IN MEDICINAL PRODUCTS
US5580578A (en) 1992-01-27 1996-12-03 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US6010715A (en) 1992-04-01 2000-01-04 Bertek, Inc. Transdermal patch incorporating a polymer film incorporated with an active agent
US6024975A (en) 1992-04-08 2000-02-15 Americare International Diagnostics, Inc. Method of transdermally administering high molecular weight drugs with a polymer skin enhancer
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
TW333456B (en) 1992-12-07 1998-06-11 Takeda Pharm Ind Co Ltd A pharmaceutical composition of sustained-release preparation the invention relates to a pharmaceutical composition of sustained-release preparation which comprises a physiologically active peptide.
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US6274552B1 (en) 1993-03-18 2001-08-14 Cytimmune Sciences, Inc. Composition and method for delivery of biologically-active factors
US5523092A (en) 1993-04-14 1996-06-04 Emory University Device for local drug delivery and methods for using the same
US5985307A (en) 1993-04-14 1999-11-16 Emory University Device and method for non-occlusive localized drug delivery
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6004534A (en) 1993-07-23 1999-12-21 Massachusetts Institute Of Technology Targeted polymerized liposomes for improved drug delivery
JPH08502759A (en) 1993-08-18 1996-03-26 アルコン ラボラトリーズ,インコーポレイテッド Use of ergoline derivatives for the treatment of glaucoma
IT1270594B (en) 1994-07-07 1997-05-07 Recordati Chem Pharm CONTROLLED RELEASE PHARMACEUTICAL COMPOSITION OF LIQUID SUSPENSION MOGUISTEIN
US5759542A (en) 1994-08-05 1998-06-02 New England Deaconess Hospital Corporation Compositions and methods for the delivery of drugs by platelets for the treatment of cardiovascular and other diseases
US5660854A (en) 1994-11-28 1997-08-26 Haynes; Duncan H Drug releasing surgical implant or dressing material
US5983134A (en) 1995-04-23 1999-11-09 Electromagnetic Bracing Systems Inc. Electrophoretic cuff apparatus drug delivery system
US6316652B1 (en) 1995-06-06 2001-11-13 Kosta Steliou Drug mitochondrial targeting agents
ATE268591T1 (en) 1995-06-27 2004-06-15 Takeda Chemical Industries Ltd METHOD FOR PRODUCING DELAYED RELEASE PREPARATIONS
US6167301A (en) 1995-08-29 2000-12-26 Flower; Ronald J. Iontophoretic drug delivery device having high-efficiency DC-to-DC energy conversion circuit
TW448055B (en) 1995-09-04 2001-08-01 Takeda Chemical Industries Ltd Method of production of sustained-release preparation
JP2909418B2 (en) 1995-09-18 1999-06-23 株式会社資生堂 Delayed release microsphere of drug
US6039975A (en) 1995-10-17 2000-03-21 Hoffman-La Roche Inc. Colon targeted delivery system
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US20010016582A1 (en) 1997-04-28 2001-08-23 Anthony H. Cincotta Method and composition for the treatment of lipid and glucose metabolism disorders
TW345603B (en) 1996-05-29 1998-11-21 Gmundner Fertigteile Gmbh A noise control device for tracks
US5877183A (en) 1996-06-06 1999-03-02 Ergo Research Corporation Treatment of lipid and glucose metabolism disorders with dopamine and serotonin agonists
JP2000513337A (en) 1996-06-06 2000-10-10 エルゴ・リサーチ・コーポレイション Treatment of lipid and glucose metabolism disorders with dopamine and serotonin agonists
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6419961B1 (en) 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
US5985317A (en) 1996-09-06 1999-11-16 Theratech, Inc. Pressure sensitive adhesive matrix patches for transdermal delivery of salts of pharmaceutical agents
EP1007012A4 (en) 1996-10-01 2006-01-18 Cima Labs Inc Taste-masked microcapsule compositions and methods of manufacture
CA2217134A1 (en) 1996-10-09 1998-04-09 Sumitomo Pharmaceuticals Co., Ltd. Sustained release formulation
ATE272394T1 (en) 1996-10-31 2004-08-15 Takeda Chemical Industries Ltd DELAYED RELEASE PREPARATION
US6131570A (en) 1998-06-30 2000-10-17 Aradigm Corporation Temperature controlling device for aerosol drug delivery
ZA9711385B (en) 1996-12-20 1999-06-18 Takeda Chemical Industries Ltd Method of producing a sustained-release preparation
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US5860957A (en) 1997-02-07 1999-01-19 Sarcos, Inc. Multipathway electronically-controlled drug delivery system
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
GB9711043D0 (en) 1997-05-29 1997-07-23 Ciba Geigy Ag Organic compounds
US5948433A (en) 1997-08-21 1999-09-07 Bertek, Inc. Transdermal patch
JP4395200B2 (en) 1997-10-28 2010-01-06 バンドー化学株式会社 Skin patch sheet and method for producing substrate sheet therefor
US6740634B1 (en) 1998-01-16 2004-05-25 Takeda Chemical Industries, Ltd. Sustained release compositions, process for producing the same and utilization thereof
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
US6048736A (en) 1998-04-29 2000-04-11 Kosak; Kenneth M. Cyclodextrin polymers for carrying and releasing drugs
KR19990085365A (en) 1998-05-16 1999-12-06 허영섭 Biodegradable polymer microspheres capable of continuously controlled controlled release and preparation method thereof
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
US6256533B1 (en) 1999-06-09 2001-07-03 The Procter & Gamble Company Apparatus and method for using an intracutaneous microneedle array
US6261595B1 (en) 2000-02-29 2001-07-17 Zars, Inc. Transdermal drug patch with attached pocket for controlled heating device
US20020123503A1 (en) 2000-12-21 2002-09-05 Malcolm Ross Cabergoline pharmaceutical compositions and methods of use thereof
JP2007505136A (en) 2003-09-10 2007-03-08 マツプ・フアーマシユーテイカルズ・インコーポレーテツド Aerosol formulation for delivering dihydroergotamine to systemic circulation by pulmonary inhalation
US20070020299A1 (en) 2003-12-31 2007-01-25 Pipkin James D Inhalant formulation containing sulfoalkyl ether cyclodextrin and corticosteroid
US9016221B2 (en) 2004-02-17 2015-04-28 University Of Florida Research Foundation, Inc. Surface topographies for non-toxic bioadhesion control
WO2006097345A1 (en) 2005-03-17 2006-09-21 Synthon Argentina S.A. Improved process for making cabergoline
GB0511060D0 (en) 2005-05-31 2005-07-06 Novartis Ag Organic compounds
DE102006013307B3 (en) 2006-03-21 2007-10-04 Ergonex Pharma Gmbh Terguride / proterguride for the treatment of chronic pain
CN104188907A (en) 2007-02-11 2014-12-10 Map药物公司 Method of therapeutic administration of DHE to enable rapid relief of migraine while minimizing side effect profile
US8859579B2 (en) 2008-03-21 2014-10-14 Richard Andrew Sewell Compostions and methods for preventing and/or treating disorders associated with cephalic pain
EP2265594A2 (en) 2008-04-16 2010-12-29 Marquette University Cysteine and cystine bioisosteres to treat schizophrenia and reduce drug cravings
RU2011140238A (en) 2009-04-15 2013-05-20 Астразенека Аб Imidazole-substituted pyrimidines useful in the treatment of diseases associated with kinase-3 glycogen synthase, such as Alzheimer's disease
IN2012DN04858A (en) 2009-12-23 2015-09-25 Map Pharmaceuticals Inc
US9867837B2 (en) * 2011-03-01 2018-01-16 Pharnext Compositions for treating neurological disorders
MX2013015373A (en) 2011-06-23 2014-02-11 Map Pharmaceuticals Inc Novel fluoroergoline analogs.

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040198753A1 (en) * 2002-01-28 2004-10-07 Hiroshi Kase Methods of treating patients suffering from movement disorders

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Golden. Dementia and Alzheimer's Disease. Minnesota Medicine January 1995 Vol. 78 pages 25-29 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11133224B2 (en) * 2019-09-27 2021-09-28 Taiwan Semiconductor Manufacturing Co., Ltd. Semiconductor structure and method for forming the same

Also Published As

Publication number Publication date
US20130345253A1 (en) 2013-12-26
US9012640B2 (en) 2015-04-21

Similar Documents

Publication Publication Date Title
US9365591B2 (en) Fluoroergoline analogs
US10308651B2 (en) Ergoline compounds and uses thereof
US8710092B2 (en) Substituted indolo 4,3 FG quinolines useful for treating migraine
US20150202198A1 (en) Novel Cabergoline Derivatives
US8946420B2 (en) Neuromodulatory compounds
US8722699B2 (en) Iso-ergoline derivatives
US9815830B2 (en) Isoergoline compounds and uses thereof
AU2012382929A1 (en) Novel cabergoline derivatives

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE