WO2010104967A1 - Sulfonamide containing compounds and uses thereof - Google Patents

Sulfonamide containing compounds and uses thereof Download PDF

Info

Publication number
WO2010104967A1
WO2010104967A1 PCT/US2010/026849 US2010026849W WO2010104967A1 WO 2010104967 A1 WO2010104967 A1 WO 2010104967A1 US 2010026849 W US2010026849 W US 2010026849W WO 2010104967 A1 WO2010104967 A1 WO 2010104967A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
ghs
subject
disorder
Prior art date
Application number
PCT/US2010/026849
Other languages
French (fr)
Inventor
Jeffrey O. Saunders
Original Assignee
Elixir 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 Elixir Pharmaceuticals, Inc. filed Critical Elixir Pharmaceuticals, Inc.
Publication of WO2010104967A1 publication Critical patent/WO2010104967A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/10Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with hetero atoms or 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 to ring carbon atoms
    • C07D261/14Nitrogen atoms

Definitions

  • the growth hormone secretagogue receptor regulates a number of physiological processes, including growth hormone (GH) release, metabolism, and appetite.
  • Ghrelin is a 28 amino acid peptide that is an endogenous ligand for the growth hormone secretagogue receptor (GHS-R) also known as the ghrelin receptor.
  • Ghrelin has been shown to stimulate feeding in humans. In rodents, ghrelin induces body weight gain and adiposity. See, e.g., Asakawa (2003) Gut 52:947. In addition to regulating feeding, ghrelin can stimulate GH secretion by activating GHS-R, particularly in somatotrophic tissue.
  • the invention features a compound of formula (I),
  • R 1 is alkoxyalkyl, alkyl, alkylC(O)NHalkyl, alkylS(O) 2 NHalkyl, alkenyl, aryl, heteroaryl, cyclyl, heterocyclyl, hydroxyalkyl, R a R b N-, R a R b Nalkyl-, R a R b Ncarboxyalkyl-, or -(CR 10 R 10 ) m R 5 ; wherein R 1 is optionally independently substituted with 1, 2, or 3 R 6 ; m is O, 1, 2, 3, or 4;
  • R is heteroaryl (e.g., a bicyclic heteroaryl such as indolyl).
  • R is substituted, for example, with 1 or 2 substituents such as halo (e.g., chloro, acyl, alkoxy, and/or alkyl (e.g., methyl or ethyl)).
  • R is cyclylalkyl
  • R is hydroxy
  • one of R or R is alkyl. In some embodiments, one of R and
  • R 5 is aryl or heteroaryl (e.g., heteroaryl such as indolyl, benzimidizolyl, or benzthiazolyl).
  • R is substituted, for example, with 1 or 2 substituents such as halo (e.g., chloro, acyl, alkoxy, and/or alkyl (e.g., methyl or ethyl)).
  • R 5 is heterocyclyl (e.g., a bicyclic heterocyclyl).
  • R 5 is 3,4-dihydro-2H-benzo[b][l,4]oxazinyl, 2,3- dihydrobenzo[b][l,4]dioxinyl, 2,3-dihydrobenzo[b][l,4]oxathiine or 2,3- dihydrobenzo[b][l,4]dithiine.
  • R 5 is substituted, for example, with 1 or 2 substituents such as halo (e.g., chloro, acyl, alkoxy, and/or alkyl (e.g., methyl or ethyl)).
  • R 5 is NR 8 C(O)R 7 .
  • R 5 is NHC(O)R 7 (e.g., NHC(O)CH 3 ).
  • R 8 is arylalkyl or heteroarylalkyl.
  • R 7 is alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl).
  • R is haloalkyl.
  • R is aryl (e.g., phenyl).
  • R 7 is cyclyl.
  • R 7 is arylalkyl (e.g., benzyl).
  • R is heteroarylalkyl. In some embodiments, R is heteroaryl (e.g., a 5 membered monocyclic heteroaryl). In some embodiments, R is substituted, for example, with 1, 2 or 3 substituents such as halo (e.g., halo (such as fluoro or chloro), acyl, alkoxy, and/or alkyl (e.g., methyl or ethyl)). In some embodiments, R is alkyl, arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl).
  • halo e.g., halo (such as fluoro or chloro)
  • acyl acyl
  • alkoxy e.g., methyl or ethyl
  • alkyl e.g., methyl or ethyl
  • R is alkyl, arylalkyl, or heteroarylalkyl (e.g.,
  • R is NR C(O)R , wherein R is alkyl (e.g., methyl), arylalkyl or heteroarylalkyl.
  • R is alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl)).
  • R is aryl (e.g., phenyl).
  • R is cyclyl.
  • R is arylalkyl (e.g., benzyl).
  • R is heteroarylalkyl.
  • R is NR C(O)NR R . In some embodiments, R is NHC(O)NR 8 R 8' . In some embodiments, R 5 is NHC(O)NHR 8' . In some embodiments, R 8 is aryl (e.g., optionally substituted, for example, with halo, alkyl or haloalkyl). In some embodiments, R is heteroaryl (e.g., a mono or bicyclic heteroaryl and/or a nitrogen containing heteroaryl (e.g., pyridinyl or pyrimidinyl)). In some embodiments, R is heteroarylalkyl (e.g., a mono or bicyclic heteroaryl).
  • R is alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl), or butyl (e.g., isobutyl)).
  • R 8 is arylalkyl.
  • R 8 is cyclyl.
  • R 8 is cyclylalkyl.
  • R 5 is NHC(O)NR 8 R 8' , wherein neither R 8 nor R 8' is H.
  • both R 8 and R 8 are alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl), or butyl (e.g., isobutyl)).
  • R 8 is cyclyl.
  • R 8 is alkyl.
  • R 8 is alkyl, arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl).
  • R and R taken together are cyclized to form -(CH2)qX(CH2) s - (e.g., a pyrrolidinyl ring or a piperidinyl ring).
  • R 5 is N(alkyl)C(O)NR 8 R 8 , N(arylalkyl)C(O)NR 8 R 8 , or N(heteroarylalkyl)C(O)NR 8 R 8 .
  • R 8 is aryl (e.g., optionally substituted, for example, with halo, alkyl or haloalkyl).
  • R is heteroaryl (e.g., a mono or bicyclic heteroaryl).
  • R is heteroarylalkyl (e.g., a mono or bicyclic heteroaryl).
  • R is alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl), or butyl (e.g., isobutyl)).
  • R is cyclylalkyl.
  • R is arylalkyl (e.g., where R is H or alkyl).
  • R is cyclyl.
  • R and R taken together are cyclized to form -(CH2) q X(CH2) s - (e.g., a pyrrolidinyl ring or a piperidinyl ring).
  • R is OC(O)NR R .
  • one of R or R is H.
  • neither R nor R is H.
  • R is aryl (e.g., optionally substituted, for example, with halo, alkyl or haloalkyl).
  • R is heteroaryl (e.g., a mono or bicyclic heteroaryl).
  • R is alkyl, arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl).
  • both R 8 and R 8 are alkyl (e.g., methyl).
  • R 5 is NR 8 C(O)OR 7 .
  • R 5 is NHC(O)OR 7 .
  • R 7 is arylalkyl (e.g., optionally substituted, for example, with alkoxy).
  • R 7 is alkyl (e.g., methyl or ethyl).
  • R 7 is cyclyl.
  • R 7 is heterocyclyl.
  • R 8 is H.
  • R 5 is NR 8 C(O)OR 7 .
  • R 8 is alkyl.
  • R 8 is arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl).
  • R is arylalkyl (e.g., optionally substituted, for example, with alkoxy).
  • R 7 is heteroarylalkyl.
  • R is alkyl (e.g., methyl or ethyl).
  • R is cyclyl.
  • R 7 is heterocyclyl.
  • R 8 is cycylalkyl (e.g., wherein R is arylalkyl).
  • R is NR C(O)SR . In some embodiments, R is NHC(O)SR . In some embodiments, R is arylalkyl such as benzyl or phenethyl (e.g., optionally substituted, for example, with alkoxy). In some embodiments, R is heteroarylalkyl. In some embodiments, R is alkyl (e.g., methyl or ethyl). In some embodiments, R is cyclyl. In some embodiments, R is heterocyclyl.
  • R is NR C(O)SR .
  • R is alkyl, arylalkyl, or heteroarylalkyl.
  • R is arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl).
  • R is arylalkyl (e.g., optionally substituted, for example, with alkoxy).
  • R is heteroarylalkyl.
  • R is alkyl (e.g., methyl or ethyl).
  • R is cyclyl. In some embodiments, R is heterocyclyl.
  • R is substituted with -NR R .
  • R and R taken together are cyclized to form -(CH2) q X(CH2) s - (e.g., a morpholino ring).
  • R is halogen, e.g., fluoro or chloro.
  • R 3 is positioned ortho to the isoxazole ring.
  • the invention features a composition comprising a compound of any of the formulae described herein and a pharmaceutically acceptable carrier.
  • the invention features a method of treating metabolic syndrome comprising administering to a subject a compound of any of the formulae described herein.
  • the invention features a method of treating obesity comprising administering to a subject a compound of any of the formulae described herein.
  • the invention features a compound that has a structure of a formula described herein, and the compound competes with ghrelin for binding to GHS-R.
  • the invention features a compound that has a structure of formula described herein, and the compound is effective for altering appetite of a subject or for altering feeding behavior of the subject.
  • a compound described herein can be administered to a subject to reduce food intake of that subject.
  • the invention features a compound that has a structure of formula described herein, and the compound is effective for modulating resistin, leptin, or adiponetin mRNA in white adipose tissue (WAT) or for modulating levels of insulin, IGF- 1, GH, Cortisol, triglycerides, free fatty acids, cholesterols (e.g., VLDL or HLDL particles) or glucose, e.g., in the blood.
  • WAT white adipose tissue
  • triglycerides free fatty acids
  • cholesterols e.g., VLDL or HLDL particles
  • glucose e.g., in the blood.
  • a compound described herein can be administered to a subject to reduce the level of cholesterol and/or triglycerides in the subject and/or reduce the ratio of total cholesterol to HDL cholesterol in a subject.
  • the invention features a compound that has a structure of formula described herein, and the compound is effective for inhibiting growth of a neoplastic cell, e.g., a cell of a ghrelin-sensitive neoplastic disorder or a GHS-R antagonist-sensitive neoplastic disorder.
  • the compound is an enantiomerically enriched isomer of a stereoisomer described herein.
  • the compound has an enantiomeric excess of at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%.
  • Enantiomer when used herein, refers to either of a pair of chemical compounds whose molecular structures have a mirror-image relationship to each other.
  • a preparation of a compound disclosed herein is enriched for an isomer of the compound having a selected stereochemistry, e.g., R or S, corresponding to a selected stereocenter, e.g., the position corresponding to the carbon alpha to the sulfonamide nitrogen in formula (I).
  • R/S configurations can be those provided in an example described herein, e.g, those described in the Table below, or the configuration of the majority or minority species in a synthetic scheme described herein.
  • the compound has a purity corresponding to a compound having a selected stereochemistry of a selected stereocenter of at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%.
  • a compound described herein includes a preparation of a compound disclosed herein that is enriched for a structure or structures having a selected stereochemistry, e.g., R or S, at a selected stereocenter, e.g., the carbon alpha to the sulfonamide nitrogen of a formula described herein e.g., formula (I).
  • a selected stereochemistry e.g., R or S
  • a selected stereocenter e.g., the carbon alpha to the sulfonamide nitrogen of a formula described herein e.g., formula (I).
  • Exemplary R/S configurations can be those provided in an example described herein, e.g, those described in the Table below, or the configuration of the majority or minority species in a synthetic scheme described herein.
  • the compound has a purity corresponding to a compound having a selected stereochemistry of a selected stereocenter of at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%.
  • a preparation of a compound disclosed herein is enriched for isomers (subject isomers) which are diastereomers of a compound described herein.
  • a compound having a selected stereochemistry, e.g., R or S, corresponding to a selected stereocenter e.g., the position corresponding to the carbon alpha to the sulfonamide nitrogen of a formula described herein e.g., formula (I).
  • R/S configurations can be those provided in an example described herein, e.g, those described in the Table below, or the configuration of the majority or minority species in a synthetic scheme described herein.
  • the invention features an organic compound that modulates (e.g., antagonizes, agonizes, or inversely agonizes) GHS-R activity, the compound having a molecular weight of less than 700 Daltons, and having fewer than four L- or D- amino acids (e.g., and any salt thereof).
  • the compound may, in certain embodiments, bind or otherwise include a metal cation.
  • the invention features a pharmaceutical composition that includes a compound described herein, e.g., a compound listed in Table 1 or described above, and a pharmaceutically acceptable carrier.
  • the invention features a method that includes: identifying a subject as having obesity, being at risk for obesity using established clinical criteria (e.g., NIH Clinical Guidelines on the Identification and Evaluation, and Treatment of Overweight and Obesity in Adults” (1998)), having insulin resistance, or being overweight; and administering a compound described herein to the subject in an amount effective to reduce weight or prevent weight gain, reduce fat content, increase metabolic activity, reduce blood glucose concentration, reduce blood insulin concentration, increase insulin sensitivity, reduce the level of cholesterol and/or triglycerides in the and/or reduce the ratio of total cholesterol to HDL cholesterol.
  • established clinical criteria e.g., NIH Clinical Guidelines on the Identification and Evaluation, and Treatment of Overweight and Obesity in Adults” (1998)
  • a compound described herein to the subject in an amount effective to reduce weight or prevent weight gain, reduce fat content, increase metabolic activity, reduce blood glucose concentration, reduce blood insulin concentration, increase insulin sensitivity, reduce the level of cholesterol and/or triglycerides in the and/or reduce the ratio of total
  • Obesity can also be defined by a subject's body mass index (BMI), which is a tool for indicating weight status, and is a measure of weight for height.
  • BMI body mass index
  • a BMI of 18.5 or below is considered underweight
  • a BMI of between 18.5 and 24.9 is considered normal
  • a BMI of between 25.0 and 29.9 is considered overweight
  • a BMI of 30.0 or greater is considered obese.
  • the BMI ranges are based on the effect body weight has on disease and death. (See World Health Organization. Physical status: The use and interpretation of anthropometry. Geneva, Switzerland: World Health Organization 1995. WHO Technical Report Series.)
  • the invention features a method of treating a subject having Prader-Willi Syndrome associated hyperphagia and obesity.
  • Prader-Willi Syndrome is a genetic disease localized to chromosome 15 that is characterized by hyperphagia, obesity, hypotonia, and mild mental retardation.
  • Obesity related mortality would include type II diabetes, cardiovascular disease, and stroke.
  • a subject having Prader-Willi Syndrome associated obesity can be identified, for example by DNA methylation test, microsatellite tests, and/or clinical phenotyping of the patient.
  • the invention features a method of treating or preventing insulin-related disorders, e.g., diabetes, retinopathy, neuropathy, nephropathy, and end organ damage.
  • the method includes administering a compound described herein, to the subject, in an amount effective to treat or prevent insulin resistance in the subject.
  • the invention features a method that includes: administering a compound described herein, to the subject, in an amount effective to reduce GHS-R activity in the subject (e.g. administering an antagonist or an inverse agonist).
  • the subject is diagnosed with or has a disorder selected from the group consisting of: cancer, diabetes, neurological disorder, obesity, age-associated disorder, neoplastic disorder, non-neoplastic disorder, cardiovascular disorder, metabolic disorder, or dermatological disorder.
  • the invention features a method of treating or preventing a disorder characterized by ghrelin levels (e.g., elevated ghrelin levels such as Prader-Willi syndrome) or GHS-R mediated signaling levels that exceed a desired or normal level.
  • ghrelin levels e.g., elevated ghrelin levels such as Prader-Willi syndrome
  • GHS-R mediated signaling levels that exceed a desired or normal level.
  • the invention features a method of treating or preventing a disorder characterized by ghrelin levels or GHS-R mediated signaling levels that are below a desired or normal level.
  • the method includes: administering a compound described herein, to a subject, in an amount effective to increase GHS-R mediating signaling in the subject, e.g., in one or more of the following tissues: pituitary, brain, spinal cord, uterus, spleen, pancreas, kidney, adrenal gland, skeletal muscle, thyroid, liver, small intestine, and heart.
  • the invention features a method of treating or preventing a GHS-R sensitive neoplastic disorder.
  • the method includes administering a compound described herein, to a subject, in an amount effective to ameliorate the neoplastic disorder (e.g., to inhibit proliferation, kill cells, or reduce or inhibit growth or an activity of neoplastic cells) in the subject.
  • the invention features a method of treating or preventing a neoplastic disorder in a subject.
  • the method includes: determining if the neoplastic disorder is mediated by cells that are sensitive to ghrelin or a GHS-R agonist or to a GHS-R antagonist, and selecting a GHS-R interacting compound described herein; and administering the selected compound to the subject.
  • the invention features a method of treating or preventing a neurodegenerative disorder.
  • the method includes: administering a compound described herein, to a subject, in an amount effective to ameliorate the neurodegenerative disorder in the subject.
  • the invention features a method of treating or preventing a metabolic disorder.
  • the method includes: administering a compound described herein, to a subject, in an amount effective to ameliorate the metabolic disorder in the subject.
  • the invention features a method of treating or preventing a cardiovascular disorder.
  • the method includes: administering a compound described herein, to a subject, in an amount effective to ameliorate the cardiovascular disorder in the subject.
  • a compound described herein can be administered to a subject to reduce the level of cholesterol and/or triglycerides in the subject and/or reduce the ratio of total cholesterol to HDL cholesterol in the subject.
  • the invention features a kit that includes a compound described herein; and instructions for administering the compound to treat a disorder described herein, e.g., an eating disorder, a metabolic disorder characterized by excess or undesired GHS-R activity, a cardiovascular disorder, a neurodegenerative disorder, and a disorder associated with altered GH/IGF-1 activity.
  • a disorder described herein e.g., an eating disorder, a metabolic disorder characterized by excess or undesired GHS-R activity, a cardiovascular disorder, a neurodegenerative disorder, and a disorder associated with altered GH/IGF-1 activity.
  • the invention features a kit that includes (1) a compound described herein; and (2) one or more reagents for monitoring expression of one or more genes regulated by GHS-R, e.g., resistin, leptin, or adiponectin, or one or more reagents for monitoring plasma levels of a metabolic regulator such as ghrelin, insulin, IGF-I or leptin.
  • GHS-R e.g., resistin, leptin, or adiponectin
  • a metabolic regulator such as ghrelin, insulin, IGF-I or leptin.
  • the invention features a method of modulating IGF-I levels (e.g., circulating IGF-I levels) in a subject.
  • the method includes administering a compound described herein.
  • a compound described herein is administered to the subject in an amount effect to modulate IGF-I levels (e.g., increase or decrease IGF-I levels).
  • antagonists are believed to be effective for decreasing IGF-I levels
  • agonists are believed to be effective for increasing IGF-I levels.
  • the invention features a method of modulating insulin levels (e.g., circulating insulin levels) in a subject.
  • the method includes administering a compound described herein.
  • a compound described herein is administered to the subject in an amount effect to modulate insulin levels (e.g., increase or decrease insulin levels).
  • antagonists are believed to be effective for decreasing insulin levels
  • agonists are believed to be effective for increasing insulin levels.
  • the invention features a method of modulating glucose levels (e.g., circulating or blood glucose levels) in a subject.
  • the method includes administering a compound described herein.
  • a compound described herein is administered to the subject in an amount effect to modulate glucose levels (e.g., increase or decrease glucose levels).
  • agonists are believed to be effective for increasing glucose levels
  • antagonists are believed to be effective for decreasing glucose levels.
  • alkylene refers to a divalent alkyl (i.e., -R-).
  • aminoalkyl refers to an alkyl substituted with an amino.
  • mercapto refers to an -SH radical.
  • thioalkoxy refers to an -S-alkyl radical.
  • alkenyl refers to a hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon double bonds.
  • the alkenyl moiety contains the indicated number of carbon atoms.
  • C2-C10 indicates that the group may have from 2 to 10 (inclusive) carbon atoms in it.
  • lower alkenyl refers to a C2-C8 alkenyl chain. In the absence of any numerical designation, “alkenyl” is a chain (straight or branched) having 2 to 10 (inclusive) carbon atoms in it.
  • alkynyl refers to a hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon triple bonds.
  • the alkynyl moiety contains the indicated number of carbon atoms.
  • C2-C10 indicates that the group may have from 2 to 10 (inclusive) carbon atoms in it.
  • lower alkynyl refers to a C2-C8 alkynyl chain. In the absence of any numerical designation, “alkynyl” is a chain (straight or branched) having 2 to 10 (inclusive) carbon atoms in it.
  • aryl refers to a 6-carbon monocyclic, 10-carbon bicyclic, or 14-carbon tricyclic ring system wherein one or more of the fused rings are aromatic, and wherein 0, 1 , 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl and the like.
  • arylalkyl or the term “aralkyl” refers to alkyl substituted with an aryl.
  • arylalkenyl refers to an alkenyl substituted with an aryl.
  • arylalkynyl refers to an alkynyl substituted with an aryl.
  • arylalkoxy refers to an alkoxy substituted with aryl.
  • cycloalkyl or "cyclyl” as employed herein includes saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12 carbons, preferably 3 to 8 carbons, and more preferably 3 to 6 carbons, wherein the cycloalkyl group may be optionally substituted.
  • Preferred cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl.
  • heteroaryl refers to an aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent.
  • heteroaryl groups include pyridyl, furyl or furanyl, imidazolyl, benzimidazolyl, pyrimidinyl, thiophenyl or thienyl, quinolinyl, indolyl, thiazolyl, and the like.
  • heteroarylalkyl or the term “heteroaralkyl” refers to an alkyl substituted with a heteroaryl.
  • heteroarylalkenyl refers to an alkenyl substituted with a heteroaryl.
  • heteroarylalkynyl refers to an alkynyl substituted with a heteroaryl.
  • heteroarylalkoxy refers to an alkoxy substituted with heteroaryl.
  • heterocyclyl or “heterocyclylalkyl” refers to a nonaromatic 5-8 membered monocyclic, 5-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent.
  • heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and include both bridged and fused ring systems.
  • heterocyclylalkyl refers to an alkyl substituted with a heterocyclyl.
  • sulfonyl refers to a sulfur attached to two oxygen atoms through double bonds.
  • alkylsulfonyl refers to an alkyl substituted with a sulfonyl.
  • GHS-R can regulate the secretion of GH.
  • GH itself is a regulator of IGF-I production.
  • compounds, e.g., compounds described herein, that modulate GHS-R activity can be used to modulate (e.g., increase or decrease) activity of the GH/IGF-1 axis.
  • agonists of GHS-R can be used to increase GH activity and/or IGF-I activity.
  • Antagonists of GHS-R can be used to decrease GH activity and/or IGF-I activity.
  • This application also incorporates by reference USSN 10/656,530, the contents of which include uses for which a compound described herein may be used, e.g., as a modulator of the GH/IGF-1 axis.
  • the GH/IGF-1 axis includes a series of extracellular and intracellular signaling components that have as a downstream target, the transcription factor Forkhead.
  • Major components of the GH/IGF-1 axis can be divided into three categories: pre-IGF 1, IGF 1, and post IGF-I components.
  • Pre IGF-I components include GH, GH-R, ghrelin, GHS- R, GHRH, GHRH-R, SST, and SST-R.
  • Post-IGF 1 components include IGF-I-R and intracellular signaling components including PI(3) kinase, PTEN phosphatase, PI(3,4)P2, 14-3-3 protein, and PI(3,4,5)P3 phosphatidyl inositol kinases, AKT serine/threonine kinase (e.g., AKT-I, AKT-2, or AKT-3), or a Forkhead transcription factor (such as FOXO-I, F0X0-3, or F0X0-4).
  • IGF-I-R intracellular signaling components
  • intracellular signaling components including PI(3) kinase, PTEN phosphatase, PI(3,4)P2, 14-3-3 protein, and PI(3,4,5)P3 phosphatidyl inositol kinases, AKT serine/threonine kinase (e.g., AKT-I, AKT-2, or AKT-3), or a Forkhead
  • a "somatotroph axis signaling pathway component” refers to a protein that is one of the following: (i) a protein that is located in a somatotroph and that regulates GH release by the somatotroph, or (ii) a protein that directly binds to a protein in class (i).
  • Exemplary somatotroph axis signaling pathway components of class (i) include cell surface receptors such as GHS-R, GHRH-R, and SST-R.
  • Exemplary somatotroph axis signaling pathway components of class (ii) include GHRH, ghrelin, and SST.
  • a compound that modulates GH levels, e.g., by altering GHS-R activity can have downstream effects.
  • the compound can alter (e.g., increase or decrease) the levels or activity of an IGF-I receptor signaling pathway effector.
  • An "IGF-I Receptor signaling pathway effector” refers a protein or other biologic whose levels are directly regulated by a Forkhead transcription factor in response to IGF 1.
  • expression of the gene encoding the protein can be directly regulated by a Forkhead transcription factor such as FOXO-I, F0X0-3a, or F0X0-4.
  • Exemplary IGF-I Receptor signaling pathway effector can include: GADD45, PA26, Selenoprotein P, Whipl, cyclin G2, and NIP3.
  • activity of the GH/IGF 1 axis refers to the net effect of the axis components with respect to ability to stimulate GH secretion, increase IGF 1 levels, or increase IGF 1 receptor signaling.
  • downstreamregulating the GH/IGF 1 axis refers to modulating one or more components such that one or more of the following is reduced, e.g., decreased GH, decreased IGF 1, or decreased IGF 1 receptor signaling. For example, in some instances, GH levels are maintained but its action is inhibited; thus IGF 1 levels are decreased without decreasing GH levels. In some instances, both GH and IGF 1 levels are decreased.
  • an "antagonist" of a particular protein includes compounds that, at the protein level, directly bind or modify the subject component such that an activity of the subject component is decreased, e.g., by competitive or non-competitive inhibition, destabilization, destruction, clearance, or otherwise.
  • the decreased activity can include reduced ability to respond to an endogenous ligand.
  • an antagonist of GHS-R can reduce the ability of GHS-R to respond to ghrelin.
  • An "agonist” of a particular protein includes compounds that, at the protein level, directly bind or modify the subject component such that an activity of the subject component is increased, e.g., by activation, stabilization, altered distribution, or otherwise.
  • An "inverse agonist" of a particular protein includes a compound that, at the protein level, causes an inhibition of the constitutive activity of the protein (e.g., a receptor), with a negative intrinsic activity, for example by binding to and/or stabilizing an inactive form of the protein, which pushes the equilibrium away from formation of an active conformation of the protein.
  • a receptor exists in an active (Ra) and an inactive (Ri) conformation.
  • Certain compounds that affect the receptor can alter the ratio of Ra to Ri (Ra/Ri).
  • a full agonist increases the ratio of Ra/Ri and can cause a "maximal", saturating effect.
  • a partial agonist when bound to the receptor, gives a response that is lower than that elicited by a full agonist (e.g., an endogenous agonist).
  • a full agonist e.g., an endogenous agonist
  • the Ra/Ri for a partial agonist is less than for a full agonist.
  • the potency of a partial agonist may be greater or less than that of the full agonist.
  • Certain compounds that agonize GHS-R to a lesser extent than ghrelin can function in assays as antagonists as well as agonists. These compounds antagonize activation of GHS-R by ghrelin because they prevent the full effect of ghrelin-receptor interaction. However, the compounds also, on their own, activate some receptor activity, typically less than a corresponding amount of ghrelin. Such compounds may be referred to as "partial agonists of GHS-R".
  • a subject with "normal" GH levels is one who would return a normal result using the glucose tolerance test in which glucose is ingested and blood levels of GH are measured by enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA) or polyclonal immunoassay.
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • polyclonal immunoassay A normal result for this test is characterized by less than 1 ng/mL of GH within 1 to 2 hours of an oral glucose load.
  • GH levels of a subject with excessive GH as in one with acromegaly may not decrease below 1 ng/mL after ingesting glucose.
  • an indicator of GH/IGF 1 axis activity refers to a detectable property of the GH/IGF 1 axis that is indicative of activity of the axis.
  • Exemplary properties include circulating GH concentration, circulating IGF 1 concentration, frequency of GH pulses, amplitude of GH pulses, GH concentration in response to glucose, IGF 1 receptor phosphorylation, and IGF 1 receptor substrate phosphorylation.
  • a compound that modulates activity of GHS-R can alter one or more indicators of GH/IGF- 1 axis activity.
  • the compounds described herein can be used for a variety of purposes, e.g., therapeutic purposes. Many of the compounds antagonize GHS-R activity and can be used to reduce GHS-R activity, e.g., in a subject. Still other compounds agonize GHS-R and can be used to increase GHS-R activity, e.g., in a subject. Some of the disclosed compounds may also provide useful biological effects by modulating the activity of cellular components other than GHS-R.
  • mice are then returned to their home cages and pre-weighed food (approximately 90 grams) is immediately returned to the food hoppers in each cage. The weight of the food remaining in the food hoppers is measured at 30 minutes, 1 hour, 2 hours, and 4 hours post compound/veh
  • Exemplary animal models for AMD include: laser-induced mouse model simulating exudative (wet) macular degeneration Bora et al., Proc. Natl. Acad. Sci. U S A., 100:2679-84 (2003); a transgenic mouse expressing a mutated form of cathepsin D resulting in features associated with the "geographic atrophy" form of AMD (Rakoczy et al., Am. J. Pathol., 161:1515-24 (2002)); and a transgenic mouse overexpressing VEGF in the retinal pigment epithelium resulting in CNV Schwesinger et al., Am. J. Pathol. 158:1161-72 (2001).
  • the compounds of the formulae described herein can, for example, be administered by injection, intravenously, intraarterially, subdermally, intraperitoneally, intramuscularly, or subcutaneously; or orally, buccally, nasally, transmucosally, topically, in an ophthalmic preparation, or by inhalation, with a dosage ranging from about 0.001 to about 100 mg/kg of body weight, e.g., between 0.001-lmg/kg, 1-lOOmg/kg, or 0.01- 5mg/kg, every 4 to 120 hours, e.g., about every 6, 8, 12, 24, 48, or 72 hours, or according to the requirements of the particular compound.
  • the methods herein contemplate administration of an effective amount of compound or compound composition to achieve the desired or stated effect (e.g., reduction of feeding in a subject).
  • the pharmaceutical compositions of this invention will be administered from about 1 to about 6 times per day, for example, the compounds can be administered about 1 to about 4 (e.g., 1, 2, 3, or 4) hours prior to meal time.
  • the compounds can be administered as a continuous infusion.
  • Such administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% active compound (w/w). Alternatively, such preparations contain from about 20% to about 80% active compound.
  • a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • the pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3- butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injec tables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms such as emulsions and or suspensions.
  • compositions of this invention may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d- ⁇ -tocopherol polyethyleneglycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-
  • the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • compositions of this invention comprise a combination of a compound of the formulae described herein and one or more additional therapeutic or prophylactic agents
  • both the compound and the additional compound should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen.
  • combinations of a plurality of compounds described herein are also envisioned.
  • the additional compounds may be administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those compounds may be part of a single dosage form, mixed together with the compounds of this invention in a single composition.
  • the term "treat” or “treatment” is defined as the application or administration of a compound, alone or in combination with, a second compound to a subject, e.g., a patient, or application or administration of the compound to an isolated tissue or cell, e.g., cell line, from a subject, e.g., a patient, who has a disorder (e.g., a disorder as described herein), a symptom of a disorder, or a predisposition toward a disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disorder, one or more symptoms of the disorder or the predisposition toward the disorder (e.g., to prevent at least one symptom of the disorder or to delay onset of at least one symptom of the disorder).
  • a disorder e.g., a disorder as described herein
  • a symptom of a disorder e.g., a disorder as described herein
  • a predisposition toward a disorder e.
  • an amount of a compound effective to treat a disorder refers to an amount of the compound which is effective, upon single or multiple dose administration to a subject, in treating a cell, or in curing, alleviating, relieving or improving a subject with a disorder beyond that expected in the absence of such treatment.
  • an amount of a compound effective to prevent a disorder refers to an amount effective, upon single- or multiple-dose administration to the subject, in preventing or delaying the occurrence of the onset or recurrence of a disorder or a symptom of the disorder.
  • the term "subject” is intended to include human and non-human animals.
  • exemplary human subjects include a human patient having a disorder, e.g., a disorder described herein or a normal subject.
  • non-human animals of the invention includes all vertebrates, e.g., non-mammals (such as chickens, amphibians, reptiles) and mammals, such as non-human primates, domesticated and/or agriculturally useful animals, e.g., sheep, dog, cat, cow, pig, etc.
  • a "metabolic disorder” is a disease or disorder characterized by an abnormality or malfunction of metabolism.
  • One category of metabolic disorders is disorders of glucose or insulin metabolism.
  • the subject can be insulin resistant, e.g., have insulin-resistance diabetes.
  • a compound described herein can be used to decrease insulin or glucose levels in a subject.
  • a compound described herein can be used to alter (e.g., increase) insulin or glucose levels in a subject.
  • Treatment with a compound may be in an amount effective to improve one or more symptoms of the metabolic disorder.
  • the invention provides a method of treating metabolic syndrome, including administering to a subject an effective amount of a compound described herein.
  • the metabolic syndrome (e.g., Syndrome X) is characterized by a group of metabolic risk factors in one person. They include: central obesity (excessive fat tissue in and around the abdomen), atherogenic dyslipidemia (blood fat disorders — mainly high triglycerides and low HDL cholesterol — that foster plaque buildups in artery walls); insulin resistance or glucose intolerance (the body can't properly use insulin or blood sugar); prothrombotic state (e.g., high fibrinogen or plasminogen activator inhibitor [-1] in the blood); raised blood pressure (i.e., hypertension) (130/85 mmHg or higher); and proinflammatory state (e.g., elevated high-sensitivity C-reactive protein in the blood).
  • central obesity excessive fat tissue in and around the abdomen
  • atherogenic dyslipidemia blood fat disorders — mainly high triglycerides and low HDL cholesterol — that foster plaque buildups in artery walls
  • insulin resistance or glucose intolerance the body can't properly use insulin or blood
  • Metabolic syndrome is closely associated with a generalized metabolic disorder called insulin resistance, in which the body can't use insulin efficiently.
  • Obesity refers to a condition in which a subject has a body mass index of greater than or equal to 30.
  • Many compounds described herein can be used to treat or prevent an over-weight condition.
  • Over-weight refers to a condition in which a subject has a body mass index of greater or equal to 25.0.
  • the body mass index (BMI) and other definitions are according to the "NIH Clinical Guidelines on the Identification and Evaluation, and Treatment of Overweight and Obesity in Adults" (1998).
  • Treatment with the compound may be in an amount effective to alter the weight of the subject, e.g., by at least 2, 5, 7, 10, 12, 15, 20, 25, 30, 25, 40, 45, 50, or 55%. Treatment with the compound may be in an amount effective to reduce the body mass index of the subject, e.g., to less than 30, 28, 27, 25, 22, 20, or 18.
  • the compounds can be used to treat or prevent aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., anorexia, bulimia, obesity, diabetes, or hyperlipidemia (e.g., elevated triglycerides and/or elevated cholesterol), as well as disorders of fat or lipid metabolism.
  • agonists of GHS-R can be used to increase food intake or to treat disorders associated with weight loss, e.g., anorexia, bulimia, and so forth.
  • Antagonists or inverse agonists of GHS-R can be used to treat aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., obesity, diabetes, or hyperlipidemia, as well as disorders of fat or lipid metabolism that results in weight gain.
  • a compound described herein is used to treat hypothalamic obesity.
  • the compound can be administered to a subject identified as at risk for hypothalamic obesity or to a subject that has an abnormal (e.g., extreme) insulin response to glucose.
  • a compound described herein can be administered to treat obesity associated with Prader-Willi Syndrome (PWS).
  • PWS is a genetic disorder associated with obesity (e.g., morbid obesity).
  • individuals suffering from PWS also have deficient GH secretion.
  • those individuals having PWS associated obesity have high fasting-ghrelin concentrations, which might contribute to hyperphagia.
  • a subject suffering from PWS associated obesity can be identified using genetic markers, determination of GH levels, fasting- ghrelin concentrations, careful phenotyping, or other methods known in the art.
  • Administration of a GHS-R antagonist such as one of the compounds described herein can be used to reduce body fat, prevent increased body fat, reduce cholesterol (e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol), and/or reduce appetite in individuals having PWS associated obesity, and/or reduce comorbidities such as diabetes, cardiovascular disease, and stroke.
  • reduce cholesterol e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol
  • a compound described herein can be administered to treat fatty liver disease (FLD), e.g., non-alcoholic fatty liver disease.
  • FLD fatty liver disease
  • FLD also known as steatorrhoeic hepatosis or steatosis hepatitis
  • FLD is a reversible condition where large vacuoles of triglyceride fat accumulate in liver cells via the process of steatosis.
  • the condition is also associated with other diseases that influence fat metabolism.
  • a "neurological disorder” is a disease or disorder characterized by an abnormality or malfunction of neuronal cells or neuronal support cells (e.g., glia or muscle).
  • the disease or disorder can affect the central and/or peripheral nervous system.
  • Exemplary neurological disorders include neuropathies, skeletal muscle atrophy, and neurodegenerative diseases, e.g., a neurodegenerative disease other than one caused at least in part by polyglutamine aggregation.
  • exemplary neurodegenerative diseases include: Alzheimer's, Amyotrophic Lateral Sclerosis (ALS), and Parkinson's disease.
  • Another class of neurodegenerative diseases includes diseases caused at least in part by aggregation of poly-glutamine.
  • Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neurological disorder.
  • a compound having GHS-R antagonist activity can be used to treat the neurological disorder.
  • a compound having, for example, GHS-R antagonist or inverse agonist activity can be used to decrease anxiety.
  • a compound having GHS-R antagonist or inverse agonist activity can be used to decrease memory retention. For example, decreasing memory retention may aid recovery from traumatic stress. In one embodiment, a compound having GHS-R agonist activity is used to increase memory retention.
  • a compound having GHS-R agonist activity is used to promote sleep in the subject or to treat sleep apnea.
  • a GHS-R agonist, inverse agonist or antagonist e.g., a compound described herein, is used to alter the circadian rhythm of a subject.
  • the compound can be delivered at particular times of day, e.g., regularly, e.g., in the evening and/or morning, to reset a circadian rhythm, e.g., prior to, during, or after traveling between time zones, or to a subject having a circadian disorder.
  • the compounds can, e.g., modulate the pulsatility of GH secretion.
  • cardiovascular disorder is a disease or disorder characterized by an abnormality or malfunction of the cardiovascular system, e.g., heart, lung, or blood vessels.
  • cardiovascular disorders include: cardiac dysrhythmias, chronic congestive heart failure, ischemic stroke, coronary artery disease and cardiomyopathy.
  • Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the cardiovascular disorder, e.g., elevated triglyceride levels or elevated cholesterol.
  • a compound having GHS-R antagonist or inverse agonist activity can be used to treat the cardiovascular disorder.
  • a “dermatological disorder” is a disease or disorder characterized by an abnormality or malfunction of the skin.
  • a “dermatological tissue condition” refers to the skin and any underlying tissue (e.g., support tissue), which contributes to the skin's function and/or appearance, e.g., cosmetic appearance. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the dermatological disorder or the dermatological tissue condition.
  • a compound having GHS-R antagonist or inverse agonist activity can be used to treat the dermatological disorder or dermatological tissue condition.
  • a "geriatric disorder” is a disease or disorder whose incidence, at the time of filing of this application and in a selected population of greater than 100,000 individuals, is at least 70% among human individuals that are greater than 70 years of age.
  • the geriatric disorder is a disorder other than cancer or a cardio-pulmonary disorder.
  • a preferred population is a United States population.
  • a population can be restricted by gender and/or ethnicity.
  • the compounds described herein can be used to treat or prevent a disorder characterized by excessive growth hormone activity.
  • the compounds can be used to reduce GH levels in the subject.
  • the subject is a human, e.g., a child (e.g., between 3-11 years), an adolescent (e.g., between 12-19 years), a young adult (e.g., between 20-25 years), or an adult.
  • a compound having GHS-R antagonist or inverse agonist activity is used to treat the disorder characterized by excessive growth hormone activity.
  • a compound described herein or other modulator of GHS-R can be administered to a subject who has a vagotomy or other disorder, which alters vagal afferent or efferent activity.
  • a subject is monitored for abnormalities in vagal nerve function, and, if a malfunction is detected, the subject is treated with a compound described herein or other modulator of GHS-R.
  • Exemplary diseases and disorders that are relevant to certain implementations include: cancer (e.g., breast cancer, colorectal cancer, CCL, CML, prostate cancer); skeletal muscle atrophy; adult-onset diabetes; diabetic nephropathy, neuropathy (e.g., sensory neuropathy, autonomic neuropathy, motor neuropathy, retinopathy); obesity; bone resorption; neurodegenerative disorders (Parkinson's disease, ALS, Alzheimer's, short- term and long-term memory loss) and disorders associated with protein aggregation (e.g., other than polyglutamine aggregation) or protein misfolding; age-related macular degeneration, Bell's Palsy; cardiovascular disorders (e.g., atherosclerosis, cardiac dysrhythmias, chronic congestive heart failure, ischemic stroke, coronary artery disease and cardiomyopathy), chronic renal failure, type 2 diabetes, ulceration, cataract, presbiopia, glomerulonephritis, Guillan-Barre syndrome, hemorrhagic stroke,
  • the compounds are directed locally to GHS-R in a target tissue of the organism.
  • GHS-R is expressed in the hypothalamus, heart, lung, pancreas, intestine, brain (particularly in the arcuate nucleus (ARC)), and adipose tissue.
  • a compound described herein can be targeted to one or more of the above tissues.
  • the compound can be formulated for inhalation for targeting to the lung.
  • the compound can be formulated for ingestion, and passage to the intestine for targeting to the intestine.
  • treatment is directed systemically, and the compound is distributed to the target tissue.
  • treatment may involve, in addition, to use of a compound in a class specified above, using a compound in another class.
  • a treatment may involve using a compound having GHS-R agonist activity.
  • treatment may involve using a compound having GHS-R antagonist activity.
  • the suitability of a particular compound can be evaluated, e.g., in an animal -based assay or by monitoring a subject.
  • NPY is a 36-amino acid peptide that stimulates food intact and depresses metabolic rate.
  • Many compounds described herein can be used to decrease NPY activity.
  • anorexigenic molecule e.g., bombesin, IL-l ⁇ , leptin, and gastrin- releasing peptide. Accordingly, the compounds may increase the discharge rate of the gastric vagal afferent.
  • GHS-R expression in human tissues is generally expressed in pituitary cells, brain, spinal cord, uterus, spleen, pancreas, kidney, adrenal gland, skeletal muscle, thyroid, liver, small intestine, and heart. Accordingly, compounds described herein can be used to treat diseases and disorders associated with undesired levels of ghrelin or ghrelin-mediated signaling activity in those tissues. For example, if the level of ghrelin or ghrelin-mediated signaling activity is undesirably low, a compound having GHS-R agonist activity can be used for treatment.
  • a compound having GHS-R antagonist activity can be used for treatment.
  • the level of desired ghrelin activity can vary from tissue to tissue. Ghrelin is secreted by the stomach and may be high in or near the stomach, but much lower in normal pancreatic tissue.
  • a "neoplastic disorder” is a disease or disorder characterized by cells that have the capacity for autonomous growth or replication, e.g., an abnormal state or condition characterized by proliferative cell growth.
  • exemplary neoplastic disorders include: carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors.
  • Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
  • a neoplastic disorder should be treated with a GHS-R agonist or antagonist can depend on the type of neoplasia. For example, Duxbury et al. (2003) Biochem. Biophys. Res. Comm. 309:464-468 report that certain neoplastic disorders are inhibited by GHS-R antagonists. These disorders include, e.g., pancreatic adenocarcinoma, and neoplasias in which GHS-R or GHS-RIb is expressed, e.g., prostate adenocarcinoma, pancreatic endocrine tumors, somatotroph tumors, and central nervous system tumors.
  • GHS-R antagonists include, e.g., pancreatic adenocarcinoma, and neoplasias in which GHS-R or GHS-RIb is expressed, e.g., prostate adenocarcinoma, pancreatic endocrine tumors,
  • neoplasia e.g., breast, lung, and thyroid adenocarcinomas can be inhibited by high levels ghrelin (> 10 nM) and, accordingly, can be treated with a GHS-R agonist, e.g., a GHS-R agonist described herein or another known GHS-R agonist.
  • GHS-R agonist e.g., a GHS-R agonist described herein or another known GHS-R agonist.
  • Neoplasias that are attenuated, inhibited, or killed by ghrelin or a GHS-R agonist are term, herein, "ghrelin-sensitive neoplastic disorders" and can be treated with a compound having GHS-R agonist activity.
  • Whether a neoplastic cell is sensitive to a ghrelin agonist or antagonist can be determined by a proliferation assay in the presence of a GHS-R agonist, e.g., ghrelin, or antagonist, e.g., D-Lys-GHRP6.
  • a proliferation assay in the presence of a GHS-R agonist, e.g., ghrelin, or antagonist, e.g., D-Lys-GHRP6.
  • Duxbury et al. disclose an exemplary proliferation assay. In one such assay, cells are seeded into 96 well plates with about 10 cells per well. The cells are cultured for 3 days in medium, and then contacted with ghrelin or D-Lys-GHRP6, or a control medium.
  • MTT assay 3-(4,5-dimethylthiazolyl-2yl)-2,5-diphenyltetrazolium) (from Trevigen, Gaithersburg, MD) for viability.
  • Other assays that can be performed are invasion and migration assays. The affect of a particular compound may also depend on concentration, which can also be varied in the assay.
  • compounds described herein can be used to treat other neoplasias and hyperplasias including "tumors," which may be benign, premalignant or malignant.
  • cancerous disorders include, but are not limited to, solid tumors, soft tissue tumors, and metastatic lesions.
  • solid tumors include malignancies, e.g., sarcomas, adenocarcinomas, and carcinomas, of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract (e.g., renal, urothelial cells), pharynx, prostate, ovary as well as adenocarcinomas which include malignancies such as most colon cancers, rectal cancer, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, cancer of the small intestine and so forth. Metastatic lesions of the aforementioned cancers can also be treated or prevented using the methods and compositions of the invention.
  • a compound described herein can be useful in treating malignancies of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract, prostate, ovary, pharynx, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus.
  • malignancies of the various organ systems such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract, prostate, ovary, pharynx, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus.
  • Exemplary solid tumors that can be treated include: fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal
  • carcinoma is recognized by those skilled in the art and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas.
  • Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary.
  • carcinosarcomas e.g., which include malignant tumors composed of carcinomatous and sarcomatous tissues.
  • An "adenocarcinoma” refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.
  • sarcoma is recognized by those skilled in the art and refers to malignant tumors of mesenchymal derivation.
  • the subject method can also be used to inhibit the proliferation of hyperplastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof.
  • the invention contemplates the treatment of various myeloid disorders including, but not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Vaickus, L. (1991) Crit Rev. in Oncol./Hemotol. 11 :267-97).
  • APML acute promyeloid leukemia
  • AML acute myelogenous leukemia
  • CML chronic myelogenous leukemia
  • Lymphoid malignancies which may be treated by the subject method, include, but are not limited to acute lymphoblastic leukemia (ALL), which includes B -lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and Waldenstrom's macroglobulinemia (WM).
  • ALL acute lymphoblastic leukemia
  • CLL chronic lymphocytic leukemia
  • PLL prolymphocytic leukemia
  • HLL hairy cell leukemia
  • WM Waldenstrom's macroglobulinemia
  • malignant lymphomas contemplated by the treatment method of the invention include, but are not limited to, non-Hodgkin's lymphoma and variants thereof, peripheral T-cell lymphomas, adult T-cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF) and Hodgkin's disease.
  • non-Hodgkin's lymphoma and variants thereof peripheral T-cell lymphomas
  • ATL adult T-cell leukemia/lymphoma
  • CTCL cutaneous T-cell lymphoma
  • LGF large granular lymphocytic leukemia
  • Hodgkin's disease Hodgkin's disease.
  • Modulators of GHS-R Compounds that modulate the activity of GHS-R can be used to treat a disorder in which a subject has less than a desired or less than a normal level of GHS-R activity, e.g., in a particular tissue; or more than a desired or higher than a normal level of GHS-R activity, e.g., in a particular tissue.
  • a metabolic disorder e.g., glucose or insulin metabolism.
  • the subject can be insulin resistant, e.g., have insulin-resistance diabetes.
  • a compound described herein can be used to decrease insulin or glucose levels in a subject.
  • a compound described herein can be used to alter (e.g., increase) insulin or glucose levels in a subject.
  • a compound described herein may be used to treat metabolic syndrome (e.g., Syndrome X), characterized by a group of metabolic risk factors in one person. They include: central obesity (excessive fat tissue in and around the abdomen), atherogenic dyslipidemia (blood fat disorders — mainly high triglycerides and low HDL cholesterol — that foster plaque buildups in artery walls); insulin resistance or glucose intolerance (the body can't properly use insulin or blood sugar); prothrombotic state (e.g., high fibrinogen or plasminogen activator inhibitor [-1] in the blood); raised blood pressure (i.e., hypertension) (130/85 mmHg or higher); and proinflammatory state (e.g., elevated high-sensitivity C-reactive protein in the blood).
  • metabolic syndrome e.g., Syndrome X
  • metabolic syndrome e.g., Syndrome X
  • the compounds described herein can be used to treat or prevent obesity.
  • the compounds can be used to treat or prevent aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., anorexia, bulimia, obesity, diabetes, or hyperlipidemia (e.g., elevated triglycerides and/or elevated cholesterol), as well as disorders of fat or lipid metabolism.
  • cardiovascular disorders include: cardiac dysrhythmias, chronic congestive heart failure, ischemic stroke, coronary artery disease and cardiomyopathy.
  • Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the cardiovascular disorder, e.g., elevated triglyceride levels or elevated cholesterol.
  • the geriatric disorder is a disorder other than cancer or a cardiopulmonary disorder.
  • a compound described herein or other modulator of GHS-R can be administered to a subject who has a vagotomy or other disorder, which alters vagal afferent or efferent activity.
  • a subject is monitored for abnormalities in vagal nerve function, and, if a malfunction is detected, the subject is treated with a compound described herein or other modulator of GHS-R.
  • Exemplary diseases and disorders that are relevant to certain implementations include: cancer (e.g., breast cancer, colorectal cancer, CCL, CML, prostate cancer); skeletal muscle atrophy; adult-onset diabetes; diabetic nephropathy, neuropathy (e.g., sensory neuropathy, autonomic neuropathy, motor neuropathy, retinopathy); obesity; bone resorption; neurodegenerative disorders (Parkinson's disease, ALS, Alzheimer's, short-term and long-term memory loss) and disorders associated with protein aggregation (e.g., other than polyglutamine aggregation) or protein misfolding; age-related macular degeneration, Bell's Palsy; cardiovascular disorders (e.g., atherosclerosis, cardiac dysrhythmias, chronic congestive heart failure, ischemic stroke, coronary artery disease and cardiomyopathy), chronic renal failure, type 2 diabetes, ulceration, cataract, presbiopia, glomerulonephritis, Guillan-Barre syndrome, hemorrhagic stroke,
  • neoplastic disorders e.g., carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors.
  • a neoplastic disorder e.g., carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors.
  • Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers.
  • Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
  • a neoplastic disorder should be treated with a GHS-R agonist or antagonist can depend on the type of neoplasia. For example, Duxbury et al. (2003) Biochem. Biophys. Res. Comm. 309:464-468 report that certain neoplastic disorders are inhibited by GHS-R antagonists. These disorders include, e.g., pancreatic adenocarcinoma, and neoplasias in which GHS-R or GHS-RIb is expressed, e.g., prostate adenocarcinoma, pancreatic endocrine tumors, somatotroph tumors, and central nervous system tumors.
  • GHS-R antagonists include, e.g., pancreatic adenocarcinoma, and neoplasias in which GHS-R or GHS-RIb is expressed, e.g., prostate adenocarcinoma, pancreatic endocrine tumors,
  • Duxbury et al. also report that certain other types of neoplasia, e.g., breast, lung, and thyroid adenocarcinomas can be inhibited by high levels ghrelin (> 10 nM) and, accordingly, can be treated with a GHS-R agonist, e.g., a GHS-R agonist described herein or another known GHS-R agonist.
  • a GHS-R agonist e.g., a GHS-R agonist described herein or another known GHS-R agonist.
  • compounds described herein can be used to treat other neoplasias and hyperplasias including tumors, which may be benign, premalignant or malignant.
  • cancerous disorders include, but are not limited to, solid tumors, soft tissue tumors, and metastatic lesions.
  • solid tumors include malignancies, e.g., sarcomas, adenocarcinomas, and carcinomas, of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract (e.g., renal, urothelial cells), pharynx, prostate, ovary as well as adenocarcinomas which include malignancies such as most colon cancers, rectal cancer, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, cancer of the small intestine and so forth.
  • Metastatic lesions of the aforementioned cancers can also be treated or prevented using the methods and compositions of the invention.
  • a compound described herein can be useful in treating malignancies of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract, prostate, ovary, pharynx, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus.
  • Exemplary solid tumors that can be treated include: fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal
  • the subject method can also be used to inhibit the proliferation of hyperplastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof.
  • the invention contemplates the treatment of various myeloid disorders including, but not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Vaickus, L. (1991) Crit Rev. in Oncol./Hemotol. 11 :267-97).
  • APML acute promyeloid leukemia
  • AML acute myelogenous leukemia
  • CML chronic myelogenous leukemia
  • Lymphoid malignancies which may be treated by the subject method, include, but are not limited to acute lymphoblastic leukemia (ALL), which includes B -lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and Waldenstrom's macroglobulinemia (WM).
  • ALL acute lymphoblastic leukemia
  • CLL chronic lymphocytic leukemia
  • PLL prolymphocytic leukemia
  • HLL hairy cell leukemia
  • WM Waldenstrom's macroglobulinemia
  • malignant lymphomas contemplated by the treatment method of the invention include, but are not limited to, non-Hodgkin's lymphoma and variants thereof, peripheral T-cell lymphomas, adult T-cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF) and Hodgkin's disease.
  • non-Hodgkin's lymphoma and variants thereof peripheral T-cell lymphomas
  • ATL adult T-cell leukemia/lymphoma
  • CTCL cutaneous T-cell lymphoma
  • LGF large granular lymphocytic leukemia
  • Hodgkin's disease Hodgkin's disease.
  • Compounds that agonize GHS-R can be used to treat a disorder in which a subject has less than a desired or less than a normal level of GHS-R activity, e.g., in a particular tissue.
  • Such compounds can be used to for one of the following purposes: to increase food intake or to treat disorders associated with weight loss, e.g., anorexia, bulimia, etc.; to increase memory retention; to promote sleep or to treat sleep apnea; treating a neoplastic disorder, e.g., e.g., breast, lung, and thyroid adenocarcinomas that may be inhibited by high levels ghrelin; treating cachexia, wasting, stimulating growth hormone release in elderly humans, patients with cancer, heart failure, or AIDS; treating growth hormone deficient adults; prevention of catabolic side effects of glucocorticoids; treatment of osteoporosis; stimulation of the immune system, acceleration of wound healing; accelerating bone fracture repair; treatment of growth retardation; treating acute or
  • Compounds that antagonize GHS-R can be used to treat a disorder in which a subject has higher than desired or higher than a normal level of GHS-R activity, e.g., in a particular tissue.
  • Antagonists or inverse agonists of GHS-R can be used to treat aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., obesity, diabetes, or hyperlipidemia, as well as disorders of fat or lipid metabolism that results in weight gain, hypothalamic obesity.
  • the compound can be administered to a subject identified as at risk for hypothalamic obesity or to a subject that has an abnormal (e.g., extreme) insulin response to glucose.
  • a compound described herein can be administered to treat obesity associated with Prader-Willi Syndrome (PWS).
  • PWS Prader-Willi Syndrome
  • Administration of a GHS-R antagonist such as one of the compounds described herein can be used to reduce body fat, prevent increased body fat, reduce cholesterol (e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol), and/or reduce appetite in individuals having PWS associated obesity, and/or reduce comorbidities such as diabetes, cardiovascular disease, and stroke.
  • exemplary neurological disorders include neuropathies, skeletal muscle atrophy, and neurodegenerative diseases, e.g., a neurodegenerative disease other than one caused at least in part by polyglutamine aggregation.
  • exemplary neurodegenerative diseases include: Alzheimer's, Amyotrophic Lateral Sclerosis (ALS), and Parkinson's disease.
  • Another class of neurodegenerative diseases includes diseases caused at least in part by aggregation of poly-glutamine.
  • Diseases of this class include: Huntington's Diseases, Spinalbulbar Muscular Atrophy (SBMA or Kennedy's Disease) Dentatorubropallidoluysian Atrophy (DRPLA), Spinocerebellar Ataxia 1 (SCAl), Spinocerebellar Ataxia 2 (SCA2), Machado-Joseph Disease (MJD; SCA3), Spinocerebellar Ataxia 6 (SCA6), Spinocerebellar Ataxia 7 (SCA7), and Spinocerebellar Ataxia 12 (SCA12).
  • DPLA Dentatorubropallidoluysian Atrophy
  • SCAl Spinocerebellar Ataxia 1
  • SCA2 Spinocerebellar Ataxia 2
  • MTD Machado-Joseph Disease
  • SCA6 Spinocerebellar Ataxia 6
  • SCA7 Spinocerebellar Ataxia 7
  • SCA12 Spinocerebellar At
  • a compound having GHS-R antagonist or inverse agonist activity can be used to decrease memory retention.
  • decreasing memory retention may aid recovery from traumatic stress.
  • AGHS-R agonist, inverse agonist or antagonist may be used to alter the circadian rhythm of a subject.
  • the compound can be delivered at particular times of day, e.g., regularly, e.g., in the evening and/or morning, to reset a circadian rhythm, e.g., prior to, during, or after traveling between time zones, or to a subject having a circadian disorder.
  • the compounds can, e.g., modulate the pulsatility of GH secretion.
  • a compound having GHS-R antagonist or inverse agonist activity can be used to treat a cardiovascular disorder.
  • a compound having GHS-R antagonist or inverse agonist activity can be used to treat fatty liver disease (e.g., non-alcoholic fatty liver disease).
  • Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the dermatological disorder or the dermatological tissue condition.
  • a compound having GHS-R antagonist or inverse agonist activity can be used to treat the dermatological disorder or dermatological tissue condition.
  • the compounds described herein can be used to treat or prevent a disorder characterized by excessive growth hormone activity.
  • the compounds can be used to reduce GH levels in the subject.
  • the subject is a human, e.g., a child (e.g., between 3-11 years), an adolescent (e.g., between 12-19 years), a young adult (e.g., between 20-25 years), or an adult.
  • a compound having GHS-R antagonist or inverse agonist activity is used to treat the disorder characterized by excessive growth hormone activity.
  • neoplastic disorder e.g., carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors.
  • a neoplastic disorder e.g., carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors.
  • Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers.
  • Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
  • GHS-R antagonists include, e.g., pancreatic adenocarcinoma, and neoplasias in which GHS-R or GHS-RIb is expressed, e.g., prostate adenocarcinoma, pancreatic endocrine tumors, somatotroph tumors, and central nervous system tumors.
  • a compound described herein may also be used to treat obesity (including inducing weight loss, reducing bodyweight, reducing food intake, reducing appetite, increasing metabolic rate, reducing fat intake, reducing carbohydrate craving, or inducing satiety); diabetes mellitus (including enhancing glucose tolerance and/or decreasing insulin resistance); type ii diabetes; hypertension; hyp erlipidcmia; osteoarthritis; cancer; gall bladder disease; sleep apnea: depression; compulsion; neuroses; insomnia/sleep disorder: substance abuse; pain: male and female sexual dysfunction (including male impotence, loss of libido, female sexual arousal dysfunction, female orgasmic dysfunction, hypoaeiive sexual desire disorder, sexual pain disorder and male erectile dysfunction); fever; inflammation; immune modulation; rheumatoid arthritis; and obesity related disorders.
  • obesity including inducing weight loss, reducing bodyweight, reducing food intake, reducing appetite, increasing metabolic rate, reducing fat intake, reducing carbohydrate craving,
  • the obesity-related disorders herein are associated with, caused by, or result from obesity.
  • F.xamples of obesity-related disorders include overeating, binge eating, and bulimia, hypertension, diabetes, elevated plasma insulin concentrations and insulin resistance, dysiipidemias, hyperlipidemia, endometrial, breast, prostate and colon cancer, osteoarthritis, obstructive sleep apnea, cholelithiasis, gallstones, heart disease, abnormal heart rhythms and arrythmias, myocardial infarction, congestive heart failure, coronary heart disease, sudden death, stroke, polycystic ovary disease, craniopharyngioma, Prader- Willi Syndrome, ProhJieh's syndrome, ( ill-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g, children with acute lymphoblastic leukemia
  • obesity-related disorders are metabolic syndrome, insulin resistance syndrome, sexual and reproductive dysfunction, such as infertility, hypogonadism in males and hirsutism in females, gastrointestinal motility disorders, such as obesity-related gastroesophageal reflux, respiratory disorders, such as ohesity-hypoventilation syndrome (Pickwickian syndrome), cardiovascular disorders, inflammation, such as systemic inflammation of the vasculature, arteriosclerosis, hypercholesterolemia, hypemrieaer ⁇ ia, lower back pain, gallbladder disease, gout, and kidney cancer, nicotine addiction, substance addiction and alcoholism.
  • metabolic syndrome such as infertility, hypogonadism in males and hirsutism in females
  • gastrointestinal motility disorders such as obesity-related gastroesophageal reflux
  • respiratory disorders such as ohesity-hypoventilation syndrome (Pickwickian syndrome)
  • cardiovascular disorders inflammation, such as systemic inflammation of the vasculature, arteriosclerosis
  • a compound described herein can be provided in a kit.
  • the kit includes (a) a composition that includes a compound described herein, and, optionally (b) informational material.
  • the informational material can be descriptive, instructional, marketing or other material that relates to the methods described herein and/or the use of the compound described herein for the methods described herein.
  • the informational material of the kits is not limited in its form.
  • the informational material can include information about production of the compound, molecular weight of the compound, concentration, date of expiration, batch or production site information, and so forth.
  • the informational material relates to use of the compound described herein to treat a disorder described herein.
  • sulfonamide 5 Boc-protected compound 4 (1 eq) will be dissolved in saturated MeOH/HCl and allowed to stir at room temperature for 1 hr. The solvent will removed under reduced pressure and the resulting deprotected amine hydrochloride salt will be washed with ether and used as such for the next step without further purification.
  • the product isoxazole amine (1.0 eq) will be suspended in DMF (40 ml) followed by the addition of triethylamine (4 eq) at 0 0 C. To this, a solution of sulfonyl chloride (2.0 eq) will be added at the same temperature and allowed to stir at room temperature for 1 hr and 30 minutes.
  • the progress of the reaction will be monitored by TLC.
  • the reaction mixture will be quenched with saturated sodium bicarbonate solution and then extracted with ethyl acetate.
  • the combined organic layer will be dried over Na 2 SO 4 , concentrated and the residue purified using silica gel column chromatography (60-120 mesh). After concentration of the column fractions, the final product will be diluted with DCM and washed with water to remove the polar impurities, concentrated and stirred with pentane to obtain the final sulfonamide 5.
  • 2,6-dichlorobenzaldehyde (25 gm, 0.143 moles, 1 eq) was dissolved in EtOH (200 ml) followed by the addition of hydroxyl amine hydrochloride (19.9 gm, 0.286 moles, 2 eq) and pyridine (34 gm, 0.429 moles, 3 eq) at 0 0 C.
  • EtOH 200 ml
  • hydroxyl amine hydrochloride (19.9 gm, 0.286 moles, 2 eq)
  • pyridine 34 gm, 0.429 moles, 3 eq
  • Oxime b (23 gm, 0.122 moles, 1 eq) was dissolved in DCM (250 ml) and to this N-chlorosuccinimide (19.6 gm, 0.1464 moles, 1.2 eq) was added at 0 0 C followed by the addition of pyridine (13.5 gm, 0.1708 moles, 1.4 eq).
  • the reaction mixture was stirred at 0 0 C for 30 minutes and monitored by TLC.
  • the reaction mass was washed with H 2 O (500 ml) and the DCM layer was separated, dried over Na 2 SO 4 , concentrated and purified immediately by fast column (silica gel, 60-120 mesh, 3-5% EA/hexane). Pale yellow solid of N-oxide c was obtained in 61% yield (14 gm) and used immediately for the next reaction. All the concentrations were done below 45 0 C and any increase in the temperature resulted in the decomposition of N-oxide.

Abstract

Compounds that modulate GHS-R are described herein.

Description

Sulfonamide containing compounds and uses thereof
CLAIM OF PRIORITY
This application claims priority from U.S.S.N. 61/158,976, filed March 10th, 2009 which is incorporated herein by reference in its entirety.
BACKGROUND
The growth hormone secretagogue receptor (GHS-R) regulates a number of physiological processes, including growth hormone (GH) release, metabolism, and appetite. Ghrelin is a 28 amino acid peptide that is an endogenous ligand for the growth hormone secretagogue receptor (GHS-R) also known as the ghrelin receptor. Ghrelin has been shown to stimulate feeding in humans. In rodents, ghrelin induces body weight gain and adiposity. See, e.g., Asakawa (2003) Gut 52:947. In addition to regulating feeding, ghrelin can stimulate GH secretion by activating GHS-R, particularly in somatotrophic tissue.
Accordingly, compounds that modulate GHS-R activity are at least useful for controlling disorders associated with GHS-R physiology.
SUMMARY
The invention relates, inter alia, to useful compounds and compositions that modulate GHS-R, as well as methods of using and making the compounds. Some examples of the compounds include sulfonamide compounds, for example heteroaryl sulfonamide compounds, and other sulfonamide compounds having cyclic moieties. Examples of heteroaryl compounds include isoxazole compounds. The compounds can be used in therapeutic applications, including modulation of disorders, diseases or disease symptoms in a subject (e.g., mammal, human, dog, cat, horse). The compounds include useful GHS-R antagonists. Such antagonists can be used, e.g., to reduce feeding in a subject.
In one aspect, the invention features a compound of formula (I),
Figure imgf000003_0001
formula (I) or a therapeutically suitable salt or prodrug thereof, wherein:
R1 is alkoxyalkyl, alkyl, alkylC(O)NHalkyl, alkylS(O)2NHalkyl, alkenyl, aryl, heteroaryl, cyclyl, heterocyclyl, hydroxyalkyl, RaRbN-, RaRbNalkyl-, RaRbNcarboxyalkyl-, or -(CR10R10 )mR5; wherein R1 is optionally independently substituted with 1, 2, or 3 R6; m is O, 1, 2, 3, or 4;
R is a member selected from the group consisting of alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, heterocycle and heterocyclealkyl, each of which is optionally substituted with -NR R ;
R3 and R4 are each members independently selected from the group consisting of hydrogen, alkyl, alkoxy, aryl, halogen, haloalkyl, cycloalkyl, cyano and nitro;
R5 is cyano, nitro, hydroxy, oxo, -OR7, -C(O)R7, -C(O)OR7, -OC(O)R7, -NR8R8', - C(O)NR8R8', -NR8C(O)R7, -NR8C(O)NR8R8', -OC(O)NR8R8', -NR8C(O)OR7, - SC(O)NR8R8', -NR8C(O)SR7, -SR9, -NR8SO2R9, -SO2NR8R8', -SOR9, and -S(O)2R9; wherein R5 is optionally substituted with 1, 2, or 3 R6 as described for R1; each R6 is independently halo, alkyl, cyclyl, heterocyclyl, aryl, heteroaryl, haloalkyl, haloalkyloxy, haloalkylthio, acyl, cyano, nitro, hydroxy, alkoxy, hydroxyalkyl, alkoxyalkyl, oxo, amino, alkylamino, dialkylamino, thiol, alkylthiol, -alkylCOOalkyl, or -alkylCOOH, CONR10R10', -C(O)alkyl, -C(O)arylalkyl;
R7 is alkyl, alkenyl, alkynyl, cyclyl, cyclylalkyl, heterocyclyl, heterocyclylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, or haloalkyl; wherein R7 may be optionally substituted as provided in R5; each R8 and R8 is independently hydrogen, acyl, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, cyclyl, or cyclylalkyl, or R and R taken together can be cyclized to form -(CH2)qX(CH2)sS wherein each R8 and R8 may independently optionally be substituted as provided in R5;
R9 is alkyl, alkenyl, alkynyl, cyclyl, cyclylalkyl, heterocyclyl, heterocyclylalkyl, aryl, arylalkyl heteroaryl, heteroarylalkyl, or haloalkyl; wherein R9 may be optionally substituted as provided in R5; q is 0, 1, 2, 3 or 4; s is 0, 1, 2, 3, or 4;
X is CR10R10', O, S, S(O), S(O)2, or NR10; each R and R is independently hydrogen or alkyl;
Ra and Rb are each members independently selected from the group consisting of hydrogen, alkoxyalkyl, alkyl, alkylcarbonyl, alkylsulfonyl, aryloxyalkyl and R0R Ncarboxyalkylcarbonyl; and
Rc and R are each independently a member selected from the group consisting of hydrogen, and alkyl.
In some embodiments, R is heteroaryl (e.g., a bicyclic heteroaryl such as indolyl). In some embodiments, R is substituted, for example, with 1 or 2 substituents such as halo (e.g., chloro, acyl, alkoxy, and/or alkyl (e.g., methyl or ethyl)).
In some embodiments, R is cyclylalkyl.
In some embodiments, R is -(CR R 2)mR • In some embodiments, one of R or R10' is alkyl (e.g., methyl). In some embodiments, R1 is -(CH2)mR5 (e.g., CH2R5).
In some embodiments, R is hydroxy.
In some embodiments, R is OR . In some embodiments, R is arylalkyl.
In some embodiments, R is NR R . In some embodiments, R and R are both
H. In some embodiments, one of R or R is alkyl. In some embodiments, one of R and
R is H. In some embodiments, R is H and R is cyclyl. In some embodiments, R is acyl.
In some embodiments, R5 is aryl or heteroaryl (e.g., heteroaryl such as indolyl, benzimidizolyl, or benzthiazolyl). In some embodiments, R is substituted, for example, with 1 or 2 substituents such as halo (e.g., chloro, acyl, alkoxy, and/or alkyl (e.g., methyl or ethyl)). In some embodiments, R5 is heterocyclyl (e.g., a bicyclic heterocyclyl). In some embodiments, R5 is 3,4-dihydro-2H-benzo[b][l,4]oxazinyl, 2,3- dihydrobenzo[b][l,4]dioxinyl, 2,3-dihydrobenzo[b][l,4]oxathiine or 2,3- dihydrobenzo[b][l,4]dithiine. In some embodiments, R5 is substituted, for example, with 1 or 2 substituents such as halo (e.g., chloro, acyl, alkoxy, and/or alkyl (e.g., methyl or ethyl)).
In some embodiments, R5 is NR8C(O)R7. In some embodiments, R5 is NHC(O)R7 (e.g., NHC(O)CH3). In some embodiments, R8 is arylalkyl or heteroarylalkyl. In some embodiments, R7 is alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl). In some embodiments, R is haloalkyl. In some embodiments, R is aryl (e.g., phenyl). In some embodiments, R7 is cyclyl. In some embodiments, R7 is arylalkyl (e.g., benzyl). In some embodiments, R is heteroarylalkyl. In some embodiments, R is heteroaryl (e.g., a 5 membered monocyclic heteroaryl). In some embodiments, R is substituted, for example, with 1, 2 or 3 substituents such as halo (e.g., halo (such as fluoro or chloro), acyl, alkoxy, and/or alkyl (e.g., methyl or ethyl)). In some embodiments, R is alkyl, arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl).
In some embodiments, R is NR C(O)R , wherein R is alkyl (e.g., methyl), arylalkyl or heteroarylalkyl. In some embodiments, R is alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl)). In some embodiments, R is aryl (e.g., phenyl). In some embodiments, R is cyclyl. In some embodiments, R is arylalkyl (e.g., benzyl). In some embodiments, R is heteroarylalkyl. In some embodiments, R is heteroaryl (e.g., a 5 or 6 membered monocyclic heteroaryl or a bicyclic heteroaryl). In some embodiments, R is substituted, for example, with 1, 2 or 3 substituents such as halo (e.g., halo (such as fluoro or chloro), acyl, alkoxy, and/or alkyl (e.g., methyl or ethyl)).
In some embodiments, R is NR C(O)NR R . In some embodiments, R is NHC(O)NR8R8'. In some embodiments, R5 is NHC(O)NHR8'. In some embodiments, R8 is aryl (e.g., optionally substituted, for example, with halo, alkyl or haloalkyl). In some embodiments, R is heteroaryl (e.g., a mono or bicyclic heteroaryl and/or a nitrogen containing heteroaryl (e.g., pyridinyl or pyrimidinyl)). In some embodiments, R is heteroarylalkyl (e.g., a mono or bicyclic heteroaryl). In some embodiments, R is alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl), or butyl (e.g., isobutyl)). In some embodiments, R8 is arylalkyl. In some embodiments, R8 is cyclyl. In some embodiments, R8 is cyclylalkyl.
In some embodiments, R5 is NHC(O)NR8R8', wherein neither R8 nor R8' is H. In some embodiments, both R8 and R8 are alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl), or butyl (e.g., isobutyl)). In some embodiments, R8 is cyclyl. In some embodiments, R8 is alkyl. In some embodiments, R8 is alkyl, arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl). In some embodiments, R and R taken together are cyclized to form -(CH2)qX(CH2)s- (e.g., a pyrrolidinyl ring or a piperidinyl ring).
In some embodiments, R5 is N(alkyl)C(O)NR8R8, N(arylalkyl)C(O)NR8R8, or N(heteroarylalkyl)C(O)NR8R8. In some embodiments, R8 is aryl (e.g., optionally substituted, for example, with halo, alkyl or haloalkyl). In some embodiments, R is heteroaryl (e.g., a mono or bicyclic heteroaryl). In some embodiments, R is heteroarylalkyl (e.g., a mono or bicyclic heteroaryl). In some embodiments, R is alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl), or butyl (e.g., isobutyl)). In some embodiments, R is cyclylalkyl. In some embodiments, R is arylalkyl (e.g., where R is H or alkyl). In some embodiments, R is cyclyl. In some embodiments, R and R taken together are cyclized to form -(CH2)qX(CH2)s- (e.g., a pyrrolidinyl ring or a piperidinyl ring).
In some embodiments, R is OC(O)NR R . In some embodiments, one of R or R is H. In some embodiments, neither R nor R is H. In some embodiments, R is aryl (e.g., optionally substituted, for example, with halo, alkyl or haloalkyl). In some embodiments, R is heteroaryl (e.g., a mono or bicyclic heteroaryl). In some embodiments, R is alkyl, arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl). In some embodiments, R is alkyl (e.g., methyl, ethyl, propyl (e.g., isopropyl), or butyl (e.g., isobutyl)). In some embodiments, R is cyclylalkyl. In some embodiments, R is arylalkyl (e.g., where R is H or alkyl). In some embodiments, R is cyclyl. In some embodiments, R8 and R8 taken together are cyclized to form - (CH2)qX(CH2)s- (e.g., a pyrrolidinyl ring or a piperidinyl ring). In some embodiments, both R8 and R8 are alkyl (e.g., methyl). In some embodiments, R5 is NR8C(O)OR7. In some embodiments, R5 is NHC(O)OR7. In some embodiments, R7 is arylalkyl (e.g., optionally substituted, for example, with alkoxy). In some embodiments, R7 is alkyl (e.g., methyl or ethyl). In some embodiments, R7 is cyclyl. In some embodiments, R7 is heterocyclyl. In some embodiments, R8 is H.
In some embodiments, R5 is NR8C(O)OR7. In some embodiments, R8 is alkyl. In some embodiments, R8 is arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl). In some embodiments, R is arylalkyl (e.g., optionally substituted, for example, with alkoxy). In some embodiments, R7 is heteroarylalkyl. In some embodiments, R is alkyl (e.g., methyl or ethyl). In some embodiments, R is cyclyl. In some embodiments, R7 is heterocyclyl. In some embodiments, R8 is cycylalkyl (e.g., wherein R is arylalkyl).
In some embodiments, R is NR C(O)SR . In some embodiments, R is NHC(O)SR . In some embodiments, R is arylalkyl such as benzyl or phenethyl (e.g., optionally substituted, for example, with alkoxy). In some embodiments, R is heteroarylalkyl. In some embodiments, R is alkyl (e.g., methyl or ethyl). In some embodiments, R is cyclyl. In some embodiments, R is heterocyclyl.
In some embodiments, R is NR C(O)SR . In some embodiments, R is alkyl, arylalkyl, or heteroarylalkyl. In some embodiments, R is arylalkyl, or heteroarylalkyl (e.g., a mono or bicyclic heteroaryl). In some embodiments, R is arylalkyl (e.g., optionally substituted, for example, with alkoxy). In some embodiments, R is heteroarylalkyl. In some embodiments, R is alkyl (e.g., methyl or ethyl). In some embodiments, R is cyclyl. In some embodiments, R is heterocyclyl.
In some embodiments, R is dialkylaminoalkyl.
In some embodiments, R is substituted with -NR R . In some embodiments, R and R taken together are cyclized to form -(CH2)qX(CH2)s- (e.g., a morpholino ring).
In some embodiments, R is halogen, e.g., fluoro or chloro. In some embodiments, R3 is positioned ortho to the isoxazole ring.
In some embodiments, R is halogen, e.g., fluoro or chloro. In some embodiments, R4 is positioned ortho to the isoxazole ring.
In some embodiments, both R and R are halogen, e.g., fluoro or chloro. In another aspect, the invention features a pharmaceutically acceptable salt comprising a compound of any of the formulae described herein.
In another aspect, the invention features a composition comprising a compound of any of the formulae described herein and a pharmaceutically acceptable carrier.
In another aspect, the invention features a method of treating metabolic syndrome comprising administering to a subject a compound of any of the formulae described herein.
In another aspect, the invention features a method of treating or delaying the onset of diabetes comprising administering to a subject a compound of any of the formulae described herein. For example, the method can include administering a compound to a subject to improve glucose tolerance, reduce fasting blood glucose, reduce post prandial blood glucose, and/or reduce HbAIc levels in a subject.
In another aspect, the invention features a method of treating obesity comprising administering to a subject a compound of any of the formulae described herein.
In one aspect, the invention features a compound that has a structure of a formula described herein, and the compound competes with ghrelin for binding to GHS-R.
In another aspect, the invention features a compound that has a structure of formula described herein, and the compound is effective for altering appetite of a subject or for altering feeding behavior of the subject. For example, a compound described herein can be administered to a subject to reduce food intake of that subject.
In another aspect, the invention features a compound that has a structure of formula described herein, and the compound is effective for modulating resistin, leptin, or adiponetin mRNA in white adipose tissue (WAT) or for modulating levels of insulin, IGF- 1, GH, Cortisol, triglycerides, free fatty acids, cholesterols (e.g., VLDL or HLDL particles) or glucose, e.g., in the blood. For example a compound described herein can be administered to a subject to reduce the level of cholesterol and/or triglycerides in the subject and/or reduce the ratio of total cholesterol to HDL cholesterol in a subject.
In another aspect, the invention features a compound that has a structure of formula described herein, and the compound is effective for inhibiting growth of a neoplastic cell, e.g., a cell of a ghrelin-sensitive neoplastic disorder or a GHS-R antagonist-sensitive neoplastic disorder. In one embodiment, the compound is an enantiomerically enriched isomer of a stereoisomer described herein. For example, the compound has an enantiomeric excess of at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%. Enantiomer, when used herein, refers to either of a pair of chemical compounds whose molecular structures have a mirror-image relationship to each other.
In one embodiment, a preparation of a compound disclosed herein is enriched for an isomer of the compound having a selected stereochemistry, e.g., R or S, corresponding to a selected stereocenter, e.g., the position corresponding to the carbon alpha to the sulfonamide nitrogen in formula (I). Exemplary R/S configurations can be those provided in an example described herein, e.g, those described in the Table below, or the configuration of the majority or minority species in a synthetic scheme described herein. For example, the compound has a purity corresponding to a compound having a selected stereochemistry of a selected stereocenter of at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%.
In one embodiment, a compound described herein includes a preparation of a compound disclosed herein that is enriched for a structure or structures having a selected stereochemistry, e.g., R or S, at a selected stereocenter, e.g., the carbon alpha to the sulfonamide nitrogen of a formula described herein e.g., formula (I).
Exemplary R/S configurations can be those provided in an example described herein, e.g, those described in the Table below, or the configuration of the majority or minority species in a synthetic scheme described herein. For example, the compound has a purity corresponding to a compound having a selected stereochemistry of a selected stereocenter of at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%.
An "enriched preparation," as used herein, is enriched for a selected stereoconfiguration of one, two, three or more selected stereocenters within the subject compound. Exemplary selected stereocenters and exemplary stereoconfigurations thereof can be selected from those provided, herein, e.g., in an example described herein, e.g., those described in the Table below. By enriched is meant at least 60%, e.g., of the molecules of compound in the preparation have a selected stereochemistry of a selected stereocenter. In preferred embodiments it is at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%. Enriched refers to the level of a subject molecule(s) and does not connote a process limitation unless specified.
In one embodiment, a preparation of a compound disclosed herein, is enriched for isomers (subject isomers) which are diastereomers of a compound described herein. For example, a compound having a selected stereochemistry, e.g., R or S, corresponding to a selected stereocenter, e.g., the position corresponding to the carbon alpha to the sulfonamide nitrogen of a formula described herein e.g., formula (I). Exemplary R/S configurations can be those provided in an example described herein, e.g, those described in the Table below, or the configuration of the majority or minority species in a synthetic scheme described herein. For example, the compound has a purity corresponding to a compound having a selected stereochemistry of a selected stereocenter of at least about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%. Diastereomer, when used herein, refers to a stereoisomer of a compound having two or more chiral centers that is not a mirror image of another stereoisomer of the same compound.
In another aspect, the invention features an organic compound that modulates (e.g., antagonizes, agonizes, or inversely agonizes) GHS-R activity, the compound having a molecular weight of less than 700 Daltons, and having fewer than four L- or D- amino acids (e.g., and any salt thereof). For example, the compound may, in certain embodiments, bind or otherwise include a metal cation.
In one embodiment, the compound has a molecular weight less than [D-Lys-3]- GHRP-6 or H(2)N-D-arg-Pro-Lys-Pro-d-Phe-Gln-d-Trp-Phe- d-Trp-Leu-Leu-NH(2) (L 756,867) or within 2, 1.5, 1.4, 1.2, 1.1, 0.8, 0.6, or 0.5 fold that of [D-Lys-3]-GHRP-6 or L 756,867.
In another aspect, the invention features a pharmaceutical composition that includes a compound described herein, e.g., a compound listed in Table 1 or described above, and a pharmaceutically acceptable carrier.
In another aspect, the invention features a method of decreasing GHS-R activity in a subject. The method includes administering the compound described herein to the subject in an amount effective to decrease GHS-R activity in the subject. In one embodiment, the subject is a mammal, e.g., a human, a primate, a dog, a cat, a racing, purebred, or an agricultural mammal. In one embodiment, the subject is overweight or obese.
In one embodiment, GHS-R activity is modulated in one or more of the following tissues: pituitary, brain, spinal cord, uterus, spleen, pancreas, kidney, adrenal gland, skeletal muscle, thyroid, liver, hypothalamus, heart, lung, pancreas, intestine, and adipose tissue.
In another aspect, the invention features a method that includes: identifying a subject as having obesity, being at risk for obesity using established clinical criteria (e.g., NIH Clinical Guidelines on the Identification and Evaluation, and Treatment of Overweight and Obesity in Adults" (1998)), having insulin resistance, or being overweight; and administering a compound described herein to the subject in an amount effective to reduce weight or prevent weight gain, reduce fat content, increase metabolic activity, reduce blood glucose concentration, reduce blood insulin concentration, increase insulin sensitivity, reduce the level of cholesterol and/or triglycerides in the and/or reduce the ratio of total cholesterol to HDL cholesterol.
Obesity can also be defined by a subject's body mass index (BMI), which is a tool for indicating weight status, and is a measure of weight for height. (See Garrow JS and Webster J. Quetelet's index (W/H2) as a measure of fatness. International Journal of Obesity 1985;9:147-153.) A BMI of 18.5 or below is considered underweight, a BMI of between 18.5 and 24.9 is considered normal, a BMI of between 25.0 and 29.9 is considered overweight, and a BMI of 30.0 or greater is considered obese. The BMI ranges are based on the effect body weight has on disease and death. (See World Health Organization. Physical status: The use and interpretation of anthropometry. Geneva, Switzerland: World Health Organization 1995. WHO Technical Report Series.) As BMI increases, the risk for some disease increases.
In another aspect, the invention features a method of treating a subject having Prader-Willi Syndrome associated hyperphagia and obesity. Prader-Willi Syndrome is a genetic disease localized to chromosome 15 that is characterized by hyperphagia, obesity, hypotonia, and mild mental retardation. (See e.g., Growth Hormone & IGF Research 13 (2003) 322-327; Growth Hormone & IGF Research 14 (2004) 1-15; The Journal of Clinical Endrocrinology & Metabolism 88(1):174-178; The Journal of Clinical Endrocrinology & Metabolism 88(5):2206-2212; The Journal of Clinical Endrocrinology & Metabolism 88(5):3573-3576; The Journal of Clinical Endrocrinology & Metabolism 87(12):5461-5464.) The method includes administering a compound described herein, to the subject, in an amount effective to maintain or reduce weight in a subject, and/or reduce appetite in a subject, control behavioral disturbances secondary to the hyperphagia, and reduce risk of morbidity and mortality associated with the extreme obesity of these individuals. Obesity related mortality would include type II diabetes, cardiovascular disease, and stroke. In some instances, a subject having Prader-Willi Syndrome associated obesity can be identified, for example by DNA methylation test, microsatellite tests, and/or clinical phenotyping of the patient.
In another aspect, the invention features a method of treating or preventing insulin-related disorders, e.g., diabetes, retinopathy, neuropathy, nephropathy, and end organ damage. The method includes administering a compound described herein, to the subject, in an amount effective to treat or prevent insulin resistance in the subject.
In another aspect, the invention features a method that includes: administering a compound described herein, to the subject, in an amount effective to reduce GHS-R activity in the subject (e.g. administering an antagonist or an inverse agonist). In one embodiment, the subject is diagnosed with or has a disorder selected from the group consisting of: cancer, diabetes, neurological disorder, obesity, age-associated disorder, neoplastic disorder, non-neoplastic disorder, cardiovascular disorder, metabolic disorder, or dermatological disorder.
For example, the compound is administered orally, or parenterally, e.g., by injection, and so forth. In one embodiment, the compound is administered at a plurality of intervals, e.g., regular intervals. In one embodiment, the method further includes monitoring the subject for GH or IGF-I activity; monitoring the subject for gene or protein regulated by GHS-R (e.g., resistin, leptin, or adiponectin) or monitoring the subject for blood or plasma levels of ghrelin, insulin, leptin and/or IGF-I.
In another aspect, the invention features a method of treating or preventing a disorder characterized by ghrelin levels (e.g., elevated ghrelin levels such as Prader-Willi syndrome) or GHS-R mediated signaling levels that exceed a desired or normal level.
The method includes: administering a compound described herein, to a subject, in an amount effective to attenuate, inhibit, or block GHS-R mediating signaling in the subject.
In another aspect, the invention features a method of treating or preventing a disorder characterized by ghrelin levels or GHS-R mediated signaling levels that are below a desired or normal level. The method includes: administering a compound described herein, to a subject, in an amount effective to increase GHS-R mediating signaling in the subject, e.g., in one or more of the following tissues: pituitary, brain, spinal cord, uterus, spleen, pancreas, kidney, adrenal gland, skeletal muscle, thyroid, liver, small intestine, and heart.
In another aspect, the invention features a method of treating or preventing a GHS-R sensitive neoplastic disorder. The method includes administering a compound described herein, to a subject, in an amount effective to ameliorate the neoplastic disorder (e.g., to inhibit proliferation, kill cells, or reduce or inhibit growth or an activity of neoplastic cells) in the subject.
In another aspect, the invention features a method of modulating feeding behavior in a subject. The method includes: administering a compound described herein, to a subject, in an amount effective to modulate a feeding behavior of the subject, e.g., to increase appetite in the subject. In one embodiment, the compound is administered prior to (e.g., at least 0.5, 1, 2, or 4 hours prior to) a mealtime or expected time at which food would be made available. In a related aspect, the method includes administering a compound, to a subject, in an amount effective to modulate a feeding behavior of the subject, e.g., to decrease appetite in the subject. In one embodiment, the compound is administered prior to (e.g., at least 0.5, 1, 2, or 4 hours prior to) a mealtime or expected time at which food would be made available.
In another aspect, the invention features a method of treating or preventing a neoplastic disorder in a subject. The method includes: determining if the neoplastic disorder is mediated by cells that are sensitive to ghrelin or a GHS-R agonist or to a GHS-R antagonist, and selecting a GHS-R interacting compound described herein; and administering the selected compound to the subject.
In another aspect, the invention features a method of treating or preventing a neurodegenerative disorder. The method includes: administering a compound described herein, to a subject, in an amount effective to ameliorate the neurodegenerative disorder in the subject.
In another aspect, the invention features a method of treating or preventing a metabolic disorder. The method includes: administering a compound described herein, to a subject, in an amount effective to ameliorate the metabolic disorder in the subject.
In another aspect, the invention features a method of treating or preventing a cardiovascular disorder. The method includes: administering a compound described herein, to a subject, in an amount effective to ameliorate the cardiovascular disorder in the subject. For example, a compound described herein can be administered to a subject to reduce the level of cholesterol and/or triglycerides in the subject and/or reduce the ratio of total cholesterol to HDL cholesterol in the subject.
In another aspect, the invention features a kit that includes a compound described herein; and instructions for administering the compound to treat a disorder described herein, e.g., an eating disorder, a metabolic disorder characterized by excess or undesired GHS-R activity, a cardiovascular disorder, a neurodegenerative disorder, and a disorder associated with altered GH/IGF-1 activity.
In another aspect, the invention features a kit that includes (1) a compound described herein; and (2) one or more reagents for monitoring expression of one or more genes regulated by GHS-R, e.g., resistin, leptin, or adiponectin, or one or more reagents for monitoring plasma levels of a metabolic regulator such as ghrelin, insulin, IGF-I or leptin.
In one aspect, the invention features a method of modulating IGF-I levels (e.g., circulating IGF-I levels) in a subject. The method includes administering a compound described herein. In one embodiment, a compound described herein is administered to the subject in an amount effect to modulate IGF-I levels (e.g., increase or decrease IGF-I levels). In particular, antagonists are believed to be effective for decreasing IGF-I levels, and agonists are believed to be effective for increasing IGF-I levels.
In one aspect, the invention features a method of modulating insulin levels (e.g., circulating insulin levels) in a subject. The method includes administering a compound described herein. In one embodiment, a compound described herein is administered to the subject in an amount effect to modulate insulin levels (e.g., increase or decrease insulin levels). In particular, antagonists are believed to be effective for decreasing insulin levels, and agonists are believed to be effective for increasing insulin levels.
In one aspect, the invention features a method of modulating glucose levels (e.g., circulating or blood glucose levels) in a subject. The method includes administering a compound described herein. In one embodiment, a compound described herein is administered to the subject in an amount effect to modulate glucose levels (e.g., increase or decrease glucose levels). In particular, agonists are believed to be effective for increasing glucose levels, and antagonists are believed to be effective for decreasing glucose levels.
The term "halo" refers to any radical of fluorine, chlorine, bromine or iodine. The term "alkyl" refers to a hydrocarbon chain that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, Cl-ClO indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it. The term "lower alkyl" refers to a C1-C8 alkyl chain. In the absence of any numerical designation, "alkyl" is a chain (straight or branched) having 1 to 10 (inclusive) carbon atoms in it. The term "alkoxy" refers to an -O-alkyl radical. The term "alkylene" refers to a divalent alkyl (i.e., -R-). The term "aminoalkyl" refers to an alkyl substituted with an amino. The term "mercapto" refers to an -SH radical. The term "thioalkoxy" refers to an -S-alkyl radical.
The term "alkenyl" refers to a hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon double bonds. The alkenyl moiety contains the indicated number of carbon atoms. For example, C2-C10 indicates that the group may have from 2 to 10 (inclusive) carbon atoms in it. The term "lower alkenyl" refers to a C2-C8 alkenyl chain. In the absence of any numerical designation, "alkenyl" is a chain (straight or branched) having 2 to 10 (inclusive) carbon atoms in it.
The term "alkynyl" refers to a hydrocarbon chain that may be a straight chain or branched chain having one or more carbon-carbon triple bonds. The alkynyl moiety contains the indicated number of carbon atoms. For example, C2-C10 indicates that the group may have from 2 to 10 (inclusive) carbon atoms in it. The term "lower alkynyl" refers to a C2-C8 alkynyl chain. In the absence of any numerical designation, "alkynyl" is a chain (straight or branched) having 2 to 10 (inclusive) carbon atoms in it. The term "aryl" refers to a 6-carbon monocyclic, 10-carbon bicyclic, or 14-carbon tricyclic ring system wherein one or more of the fused rings are aromatic, and wherein 0, 1 , 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl and the like. The term "arylalkyl" or the term "aralkyl" refers to alkyl substituted with an aryl. The term "arylalkenyl" refers to an alkenyl substituted with an aryl. The term "arylalkynyl" refers to an alkynyl substituted with an aryl. The term "arylalkoxy" refers to an alkoxy substituted with aryl.
The terms "cycloalkyl" or "cyclyl" as employed herein includes saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12 carbons, preferably 3 to 8 carbons, and more preferably 3 to 6 carbons, wherein the cycloalkyl group may be optionally substituted. Preferred cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl.
The term "heteroaryl" refers to an aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2, 3, or 4 atoms of each ring may be substituted by a substituent. Examples of heteroaryl groups include pyridyl, furyl or furanyl, imidazolyl, benzimidazolyl, pyrimidinyl, thiophenyl or thienyl, quinolinyl, indolyl, thiazolyl, and the like. The term "heteroarylalkyl" or the term "heteroaralkyl" refers to an alkyl substituted with a heteroaryl. The term "heteroarylalkenyl" refers to an alkenyl substituted with a heteroaryl. The term "heteroarylalkynyl" refers to an alkynyl substituted with a heteroaryl. The term "heteroarylalkoxy" refers to an alkoxy substituted with heteroaryl.
The term "heterocyclyl" or "heterocyclylalkyl" refers to a nonaromatic 5-8 membered monocyclic, 5-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein 0, 1, 2 or 3 atoms of each ring may be substituted by a substituent. Examples of heterocyclyl groups include piperazinyl, pyrrolidinyl, dioxanyl, morpholinyl, tetrahydrofuranyl, and include both bridged and fused ring systems. The term "heterocyclylalkyl" refers to an alkyl substituted with a heterocyclyl.
The term "sulfonyl" refers to a sulfur attached to two oxygen atoms through double bonds. An "alkylsulfonyl" refers to an alkyl substituted with a sulfonyl.
The term "substituents" refers to a group "substituted" on an alkyl, cycloalkyl, aryl, heterocyclyl, or heteroaryl group at any atom of that group. Any moiety described herein can be further substituted with a substituent. Suitable substituents include, without limitation, halo, hydroxy, mercapto, oxo, nitro, haloalkyl, alkyl, aryl, aralkyl, alkoxy, thioalkoxy, aryloxy, amino, alkoxycarbonyl, amido, carboxy, alkanesulfonyl, alkylcarbonyl, and cyano groups.
GHS-R can regulate the secretion of GH. GH itself is a regulator of IGF-I production. Thus, compounds, e.g., compounds described herein, that modulate GHS-R activity can be used to modulate (e.g., increase or decrease) activity of the GH/IGF-1 axis. For example, agonists of GHS-R can be used to increase GH activity and/or IGF-I activity. Antagonists of GHS-R can be used to decrease GH activity and/or IGF-I activity. This application also incorporates by reference USSN 10/656,530, the contents of which include uses for which a compound described herein may be used, e.g., as a modulator of the GH/IGF-1 axis.
The GH/IGF-1 axis includes a series of extracellular and intracellular signaling components that have as a downstream target, the transcription factor Forkhead. Major components of the GH/IGF-1 axis can be divided into three categories: pre-IGF 1, IGF 1, and post IGF-I components. "Pre IGF-I components" include GH, GH-R, ghrelin, GHS- R, GHRH, GHRH-R, SST, and SST-R. "Post-IGF 1 components" include IGF-I-R and intracellular signaling components including PI(3) kinase, PTEN phosphatase, PI(3,4)P2, 14-3-3 protein, and PI(3,4,5)P3 phosphatidyl inositol kinases, AKT serine/threonine kinase (e.g., AKT-I, AKT-2, or AKT-3), or a Forkhead transcription factor (such as FOXO-I, F0X0-3, or F0X0-4). A "somatotroph axis signaling pathway component" refers to a protein that is one of the following: (i) a protein that is located in a somatotroph and that regulates GH release by the somatotroph, or (ii) a protein that directly binds to a protein in class (i). Exemplary somatotroph axis signaling pathway components of class (i) include cell surface receptors such as GHS-R, GHRH-R, and SST-R. Exemplary somatotroph axis signaling pathway components of class (ii) include GHRH, ghrelin, and SST.
A compound that modulates GH levels, e.g., by altering GHS-R activity can have downstream effects. For example, the compound can alter (e.g., increase or decrease) the levels or activity of an IGF-I receptor signaling pathway effector. An "IGF-I Receptor signaling pathway effector" refers a protein or other biologic whose levels are directly regulated by a Forkhead transcription factor in response to IGF 1. For example, expression of the gene encoding the protein can be directly regulated by a Forkhead transcription factor such as FOXO-I, F0X0-3a, or F0X0-4. Exemplary IGF-I Receptor signaling pathway effector can include: GADD45, PA26, Selenoprotein P, Whipl, cyclin G2, and NIP3.
As used herein, "activity of the GH/IGF 1 axis" refers to the net effect of the axis components with respect to ability to stimulate GH secretion, increase IGF 1 levels, or increase IGF 1 receptor signaling. Accordingly, "downregulating the GH/IGF 1 axis" refers to modulating one or more components such that one or more of the following is reduced, e.g., decreased GH, decreased IGF 1, or decreased IGF 1 receptor signaling. For example, in some instances, GH levels are maintained but its action is inhibited; thus IGF 1 levels are decreased without decreasing GH levels. In some instances, both GH and IGF 1 levels are decreased.
An "antagonist" of a particular protein includes compounds that, at the protein level, directly bind or modify the subject component such that an activity of the subject component is decreased, e.g., by competitive or non-competitive inhibition, destabilization, destruction, clearance, or otherwise. For example, the decreased activity can include reduced ability to respond to an endogenous ligand. For example, an antagonist of GHS-R can reduce the ability of GHS-R to respond to ghrelin.
An "agonist" of a particular protein includes compounds that, at the protein level, directly bind or modify the subject component such that an activity of the subject component is increased, e.g., by activation, stabilization, altered distribution, or otherwise. An "inverse agonist" of a particular protein includes a compound that, at the protein level, causes an inhibition of the constitutive activity of the protein (e.g., a receptor), with a negative intrinsic activity, for example by binding to and/or stabilizing an inactive form of the protein, which pushes the equilibrium away from formation of an active conformation of the protein.
Generally, a receptor exists in an active (Ra) and an inactive (Ri) conformation. Certain compounds that affect the receptor can alter the ratio of Ra to Ri (Ra/Ri). For example, a full agonist increases the ratio of Ra/Ri and can cause a "maximal", saturating effect. A partial agonist, when bound to the receptor, gives a response that is lower than that elicited by a full agonist (e.g., an endogenous agonist). Thus, the Ra/Ri for a partial agonist is less than for a full agonist. However, the potency of a partial agonist may be greater or less than that of the full agonist.
Certain compounds that agonize GHS-R to a lesser extent than ghrelin can function in assays as antagonists as well as agonists. These compounds antagonize activation of GHS-R by ghrelin because they prevent the full effect of ghrelin-receptor interaction. However, the compounds also, on their own, activate some receptor activity, typically less than a corresponding amount of ghrelin. Such compounds may be referred to as "partial agonists of GHS-R".
A subject with "normal" GH levels is one who would return a normal result using the glucose tolerance test in which glucose is ingested and blood levels of GH are measured by enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA) or polyclonal immunoassay. A normal result for this test is characterized by less than 1 ng/mL of GH within 1 to 2 hours of an oral glucose load. However, GH levels of a subject with excessive GH, as in one with acromegaly may not decrease below 1 ng/mL after ingesting glucose. Because GH levels oscillate every twenty to thirty minutes and varies in level according to the time of day, stress level, exercise, etc., a standard means of determining if GH levels are excessive is to administer glucose. This approach normalizes GH and is less affected by the pulsatility of GH, age, gender, or other factors. Alternatively or as a confirmation, since IGF 1 levels are invariably increased in acromegalic individuals, IGF 1 levels can be measured and compared to age and gender matched normal controls. The term "an indicator of GH/IGF 1 axis activity" refers to a detectable property of the GH/IGF 1 axis that is indicative of activity of the axis. Exemplary properties include circulating GH concentration, circulating IGF 1 concentration, frequency of GH pulses, amplitude of GH pulses, GH concentration in response to glucose, IGF 1 receptor phosphorylation, and IGF 1 receptor substrate phosphorylation. A compound that modulates activity of GHS-R can alter one or more indicators of GH/IGF- 1 axis activity.
The details of one or more embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and from the claims.
DETAILED DESCRIPTION
The compounds described herein can be used for a variety of purposes, e.g., therapeutic purposes. Many of the compounds antagonize GHS-R activity and can be used to reduce GHS-R activity, e.g., in a subject. Still other compounds agonize GHS-R and can be used to increase GHS-R activity, e.g., in a subject. Some of the disclosed compounds may also provide useful biological effects by modulating the activity of cellular components other than GHS-R.
Compounds of formula (I) are described herein, for example as provided in the summary above. Exemplary compounds are shown in Table 1. Table 1
Figure imgf000020_0001
Figure imgf000021_0001
Other aspects of this invention relate to a composition having a compound of any of the formulae described herein and a pharmaceutically acceptable carrier; or a compound of any of the formulae described herein, an additional therapeutic compound (e.g., an anti-hypertensive compound or a cholesterol lowering compound), and a pharmaceutically acceptable carrier; or a compound of any of the formulae described herein, an additional therapeutic compound, and a pharmaceutically acceptable carrier.
Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term "stable", as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject).
Synthesis of Ghrelin Receptor-Modulating Compounds
The compounds described herein can be made using a variety of synthetic techniques. Exemplary methods for making sulfonamide compounds include those described in U.S. 20070270473, the contents of which are incorporated herein by reference.
Scheme A below depicts a representative synthesis of certain compounds described herein. Scheme A.
1 ester hydrolysis
2 Curtius rearrangement
3 deprotection
Figure imgf000022_0001
Sub = substituent LG = leaving group R2, R3 & R4 = groups as
Figure imgf000022_0002
defined herein
As can be appreciated by the skilled artisan, further methods of synthesizing the compounds of the formulae herein will be evident to those of ordinary skill in the art. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents or Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof. Additionally, the compounds disclosed herein can be prepared on a solid support.
The term "solid support" refers a material to which a compound is attached to facilitate identification, isolation, purification, or chemical reaction selectivity of the compound. Such materials are known in the art and include, for example, beads, pellets, disks, fibers, gels, or particles such as cellulose beads, pore-glass beads, silica gels, polystyrene beads optionally cross-linked with divinylbenzene and optionally grafted with polyethylene glycol, poly-acrylamide beads, latex beads, dimethylacrylamide beads optionally cross-linked with N,N'-bis-acryloyl ethylene diamine, glass particles coated with hydrophobic polymer, and material having a rigid or semi-rigid surface. The solid supports optionally have functional groups such as amino, hydroxy, carboxy, or halo groups, (see, Obrecht, D. and Villalgrodo, J.M., Solid-Supported Combinatorial and Parallel Synthesis of Small-Molecular-Weight Compound Libraries, Pergamon-Elsevier Science Limited (1998)), and include those useful in techniques such as the "split and pool" or "parallel" synthesis techniques, solid-phase and solution-phase techniques, and encoding techniques (see, for example, Czarnik, A. W., Curr. Opin. Chem. Bio., (1997) 1, 60).
The compounds of this invention may contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers and enantiometric mixtures, individual diastereomers and diastereomeric mixtures. All such isomeric forms of these compounds are expressly included in the present invention. The compounds of this invention may also be represented in multiple tautomeric forms, in such instances, the invention expressly includes all tautomeric forms of the compounds described herein (e.g., alkylation of a ring system may result in alkylation at multiple sites, the invention expressly includes all such reaction products). All such isomeric forms of such compounds are expressly included in the present invention. All crystal forms of the compounds described herein are expressly included in the present invention.
As used herein, the compounds of this invention, including the compounds of formulae described herein, are defined to include pharmaceutically acceptable derivatives or prodrugs thereof. A "pharmaceutically acceptable derivative or prodrug" means any pharmaceutically acceptable salt, ester, salt of an ester, or other derivative of a compound of this invention (for example an imidate ester of an amide), which, upon administration to a recipient, is capable of providing (directly or indirectly) a compound of this invention. Particularly favored derivatives and prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species. Preferred prodrugs include derivatives where a group which enhances aqueous solubility or active transport through the gut membrane is appended to the structure of formulae described herein. The compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acid salts include acetate, adipate, benzoate, benzenesulfonate, butyrate, citrate, digluconate, dodecylsulfate, formate, fumarate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, palmoate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, tartrate, tosylate and undecanoate. Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N-(alkyl)4 + salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization.
Evaluating Compounds
A variety of methods can be used to evaluate a compound for ability to modulate GHS-R activity. Evaluation methods include in vitro binding assays, in vitro cell-based signaling assays, and in vivo methods. The evaluation methods can evaluate binding activity, or an activity downstream of GHS-R, e.g., a signaling activity downstream of GHS-R such as inositol phosphate production, Ca + mobilization, or gene transcription (e.g., CREB-mediated gene transcription).
Binding assays. Generally, the compounds can be evaluated to determine if they bind to GHS-R and if they compete with one or more known compounds that interact with GHS-R, and the extent of such interactions. For example, the compounds can be evaluated to determine if they compete with ghrelin, ipamorelin, L-692,400 or L- 692,492. One exemplary binding assay is as follows: GHS-R expressing COS-7 cells cultured at a density of 1 x 105 cells per well so that binding is assayed in the range of about 5 - 8 % binding of the radioactive ligand. For example, the cells can express an endogenous nucleic acid encoding GHS-R or an exogenous nucleic acid encoding GHS- R. Cells transfected with an exogenous nucleic acid encoding GHS-R can be used, e.g., two days, after transfection. Competition binding experiments are performed for 3 hours at 4°C using 25 pM of 125I-ghrelin in 0.5 ml of 50 mM HEPES buffer, pH 7.4, supplemented with 1 mM CaCl2, 5 mM MgCl2, and 0.1 % (w/v) bovine serum albumin, 40 mg/ml bacitracin. Non-specific binding can be determined as the binding in the presence of 1 mM of unlabeled ghrelin. Cells are washed twice in 0.5 ml of ice-cold buffer and then lysed with 0.5-1 ml of lysis buffer (8 M Urea, 2 % NP40 in 3 M acetic acid). After washing and lysis, the bound radioactivity is counted. Assays can be run in duplicate or triplicate, e.g., to provide statistical power.
Values of the dissociation and inhibition constants (Kd and K1) can be estimated from competition binding experiments using the equation:
Kd = IC50-Z, and K1 = IC50 / (I + L I Kd), where L is the concentration of radioactive ligand. Bmax values can be estimated from competition binding experiments using the equation Bmax = Bo ICso/[ligand] , where Bo is the specifically bound radioligand.
Cell-Based Activity Assays. For example, the ability of the compound to modulate accumulation of a second messenger signaling component downstream of GHS-R can be evaluated. For example, inositol phosphates (IP), as a result of Gq signaling in a mammalian cell, e.g., a Cos-7 cells. Other tissue culture cells, Xenopus oocytes, and primary cells can also be used.
Phosphatidylinositol turnover assay. One day after transfection COS-7 cells are incubated for 24 hours with 5 μCi of [3H] -myoinositol in 1 ml medium supplemented with 10% fetal calf serum, 2 mM glutamine and 0.01 mg/ml gentamicin per well. Cells are then washed twice in buffer, 20 mM HEPES, pH 7.4, supplemented with 140 mM NaCl, 5 mM KCl, 1 mM MgSO4, 1 mM CaCl2, 10 mM glucose, 0.05 % (w/v) bovine serum; and incubated in 0.5 ml buffer supplemented with 10 mM LiCl at 37°C for 30 min. For some assays, it is also useful to incubate the cells with adenosine deaminase ADA (200U/mg, Boehringer Mannheim, Germany) for 30 min in a concentration of lU/ml .
After incubation with the compound of interest for 45 min at 37°C, cells are extracted with 10 % ice-cold perchloric acid and placed on ice for 30 min. The resulting supernatants are neutralized with KOH in HEPES buffer, and [3H] -inositol phosphate is purified on Bio-Rad AG 1-X8 anion exchange resin as described. Assays can be run in duplicate, triplicate, etc.
Other second messenger assays. Another second messenger that can be evaluated is Ca2+. Ca2+ mobilization can be evaluated using a calcium sensitive detector, such as aequorin protein or a dye, e.g., FURA-2. In an exemplary assay, calcium mobilization is evaluated in a recombinant cell that expresses GHS-R and aequorin. Gene expression assay. HEK293 cells (30 000 cells/well) seeded in 96-well plates are transiently transfected with a mixture of pFA2-CREB and pFR-Luc reporter plasmid (PathDetect™ CREB trans-Reporting System, Stratagene) and nucleic acid encoding GHS. One day after trans fection, cells are treated with the compound of interest in an assay volume of 100 μl medium for 5 hrs. After treatment, cells are cultured in low serum (2.5%). After the incubation period, the assay is ended by washing the cells twice with PBS and adding lOOμl luciferase assay reagent (LucLite™, Packard Bioscience). Luminescence is measured (e.g., as relative light units (RLU)) using in a luminometer such as the TopCounter™ (Packard Bioscience) for 5 sec.
Other transcription based assays can include evaluating transcription of GHS-R regulated genes in primary cells that express GHS-R (e.g., cells from pituitary, brain, spinal cord, uterus, spleen, pancreas, kidney, adrenal gland, skeletal muscle, thyroid, liver, small intestine, and heart) or in recombinant cells that express GHS-R. mRNA levels can be evaluated by any method, e.g., microarray analysis, Northern blotting, or RT-PCR. Exemplary genes that are directly or indirectly regulated by GHS-R activity include leptin, resistin, and adiponectin. GHS-R activity may also affect insulin, IGF-I, and leptin levels in circulation.
IC50 and EC50 values can be determined by nonlinear regression, e.g., using the Prism 3.0 software (GraphPad Software, San Diego). In vivo assays. Exemplary in vivo assays include the fast-refeeding assay described in Example 1 and as follows.
Prior to compound administration, mice are weighed and sorted into groups based on comparable body weight. Food is removed at 6pm for an overnight (- 16 hour) fast. Beginning at 10 am on the next morning, mice are administered with either vehicle (e.g., saline + acetic acid, pH=5) or the compound of interest. Mice are then returned to their home cages and pre-weighed food (approximately 90 grams) is immediately returned to the food hoppers in each cage. The weight of the food remaining in the food hoppers is measured at 30 minutes, 1 hour, 2 hours, and 4 hours post compound/vehicle administration. Final body weights are then recorded for the mice.
The compound of interest can also be evaluated in other experiments. For example, the compound can be administered to lean or obese mice (e.g,. (ob/ob) C57BL/6J mice), or other experimental animals. The compound can be administered intraperitoneally or intracerebroventricularly. After administration, the animal is evaluated, e.g., for feeding behavior, anxiety, or one or more physiological parameters, e.g., a metabolic parameter.
ICV Administration. For intra-third cerebroventricular (ICV) administration, each drug can be diluted in 4 Tl of artificial cerebrospinal fluid for injection. For ICV injection, mice are anaesthetized with sodium pentobarbital (80-85 mg/kg intraperitoneally) and placed in a stereotaxic instrument seven days before the experiments. A hole is made in each skull using a needle inserted 0.9 mm lateral to the central suture and 0.9 mm posterior to the bregma. A 24 gauge cannula bevelled at one end over a distance of 3 mm is implanted into the third cerebral ventricle for ICV injection.
Gastric emptying assessment. Another test for food consumption after administration of a compound of interest is the gastric emptying assessment. Before the gastric emptying assessment, mice are food deprived for 16 hours with free access to water. Fasted mice are given free access to preweighed pellets for one hour and then administered the compound of interest. The mice are again deprived of food for one or two hours after the compound administration. Food intake is measured by weighing uneaten pellets. Mice are killed by cervical dislocation two or three hours after the compound administration. Immediately after the stomach was exposed by laparotomy, quickly ligated at both the pylorus and cardia, removed, and the dry content is weighed. Gastric emptying is calculated according to the following formula: gastric emptying (%) = (1 - (dry weight of food recovered from the stomach/weight of food intake)) x 100.
Anxiety tests. Anxiety can be assessed in the standard elevated plus maze, 50 cm above the ground. The four arms can be made 27 cm long and 6 cm wide. Two opposing arms are enclosed by walls 15 cm high (closed arms) while the other arms are devoid of walls (open arms). Each mouse is placed in the center of the maze facing one of the enclosed arms 10 minutes after injection with a compound. The cumulative time spent in each arm and the number of entries into the open or closed arms is recorded during a five minute test session. The time spent in the open arms is expressed as a percentage of total entry time (100-open/ open+closed) and the number of entries in the open arms is expressed as a percentage of the total number of entries (100-open/total entries).
Parameter analysis. Mice or other animals provided with the test compound can be analyzed for one or more biological parameters, e.g., metabolic parameters. For mice, serum is obtained from blood from the orbital sinus under ether anesthesia at the end of a treatment (e.g., eight hours after removal of food and the final intraperitoneal injection). Mice are killed by cervical dislocation. Immediately after, the epididymal fat pad mass can be assessed based on removal and weighing of the white adipose tissue (WAT) and the gastrocnemius muscle. Blood glucose can be measured by the glucose oxidase method. Serum insulin and free fatty acids (FFA) can be measured by enzyme immunoassay and an enzymatic method (Eiken Chemical Co., Ltd, Tokyo, Japan), respectively. Serum triglycerides and total cholesterol can be measured by an enzymatic method (Wako Pure Chemical Industries, Ltd, Tokyo, Japan). mRNA analysis. RNA is isolated from the stomach, epididymal fat or other relevant tissues using the RNeasy Mini Kit (Qiagen, Tokyo, Japan). Total RNA is denatured with formaldehyde, electrophoresed in 1% agarose gel, and blotted onto a Hybond N+ membrane. The membranes are hybridized with a labeled cDNAprobe (e.g., radioactively, chemically, or fluorescently labeled) for the gene of interest. The total integrated densities of hybridization signals can be determined by densitometry. Data can be normalized to a glycer aldehyde 3 -phosphate dehydrogenase mRNA abundance or to actin mRNA abundance and expressed as a percentage of controls. Exemplary genes that can be evaluated include ghrelin, leptin, resistin, and adiponectin. It is also possible to use a transgenic animal that includes a reporter construct with a regulatory region from the gene of interest or to use a recombinant cell with such a construct.
A compound described herein can have a K1 (as an antagonist) of less than 200, 100, 80, 70, 60, or 50 nM, in one or more of the described assays. A compound described herein can have a KD as an agonist of greater than 20, 40, 50, 100, 200, 300, or 500 nM, in one or more of the described assays.
A compound described herein can also specifically interact with GHS-R, e.g., relative to other cell surface receptors. The motilin receptor, for example, is a homolog of GHS-R. A disclosed compound may preferentially interact with GHS-R relative to the motilin receptor, e.g., at least a 2, 5, 10, 20, 50, or 100 preference. In another embodiment, the disclosed compound may also interact with motilin receptor, and, e.g., alter motilin receptor activity.
In one embodiment, the compound may alter an intracellular signaling activity downstream of GHS-R, e.g., Gq signaling, phospholipase C signaling, and cAMP response element (CRE) driven gene transcription.
Compounds may also be evaluated for their therapeutic activity with respect to any disorder, e.g., a disorder described herein. Animal models for many disorders are well known in the art.
Cells and animals for evaluating the effect of a compound on ALS status include a mouse which has an altered SOD gene, e.g., a SOD1-G93A transgenic mouse which carries a variable number of copies of the human G93A SOD mutation driven by the endogenous promoter, a SOD1-G37R transgenic mouse (Wong et al., Neuron, 14(6): 1105-16 (1995)); SOD1-G85R transgenic mouse (Bruijn et al., Neuron, 18(2):327- 38 (1997)); C. elegans strains expressing mutant human SODl (Oeda et al., Hum MoI Genet., 10:2013-23 (2001)); and a Drosophila expressing mutations in Cu/Zn superoxide dismutase (SOD). (Phillips et al., Proc. Natl. Acad. Sci. U.S.A., 92:8574-78 (1995) and McCabe, Proc. Natl. Acad. Sci. U.S.A., 92:8533-34 (1995)).
Cells and animals for evaluating the effect of a compound on Alzheimer's disease are described, e.g., in US 6,509,515 and US 5,387,742; 5,877,399; 6,358,752; and 6,187,992. In US 6,509,515, the animal expresses an amyloid precursor protein (APP) sequence at a level in brain tissues such that the animal develops a progressive neurologic disorder. An exemplary animal model for evaluating polyglutamine-based aggregation is the transgenic mouse strain is the R6/2 line (Mangiarini et al. Cell 87: 493-506 (1996)). Models for evaluating the effect of a test compound on muscle atrophy include, e.g.,: 1) rat medial gastrocnemius muscle mass loss resulting from denervation, e.g., by severing the right sciatic nerve at mid-thigh; 2) rat medial gastrocnemius muscle mass loss resulting from immobilization, e.g., by fixed the right ankle joint at 90 degrees of flexion; 3) rat medial gastrocnemius muscle mass loss resulting from hindlimb suspension; (see, e.g., U.S. 2003-0129686); 4) skeletal muscle atrophy resulting from treatment with the cachectic cytokine, interleukin-1 (IL-I) (R. N. Cooney, S. R. Kimball, T. C. Vary, Shock 7, 1-16 (1997)); and 5) skeletal muscle atrophy resulting from treatment with the glucocorticoid, dexamethasone (A. L. Goldberg, J Biol Chem 244, 3223-9 (1969).). Models 1 , 2, and 3 induce muscle atrophy by altering the neural activity and/or external load a muscle experiences to various degrees. Models 4 and 5 induce atrophy without directly affecting those parameters.
Exemplary animal models for AMD (age-related macular degeneration) include: laser-induced mouse model simulating exudative (wet) macular degeneration Bora et al., Proc. Natl. Acad. Sci. U S A., 100:2679-84 (2003); a transgenic mouse expressing a mutated form of cathepsin D resulting in features associated with the "geographic atrophy" form of AMD (Rakoczy et al., Am. J. Pathol., 161:1515-24 (2002)); and a transgenic mouse overexpressing VEGF in the retinal pigment epithelium resulting in CNV Schwesinger et al., Am. J. Pathol. 158:1161-72 (2001).
Exemplary animal models of Parkinson's disease include primates rendered parkinsonian by treatment with the dopaminergic neurotoxin l-methyl-4 phenyl 1,2,3,6- tetrahydropyridine (MPTP) (see, e.g., US Appl 20030055231 and Wichmann et al., Ann. N.Y. Acad. Sci., 991:199-213 (2003); 6-hydroxydopamine-lesioned rats (e.g., Lab. Anim. Sci. ,49:363-71 (1999)); and transgenic invertebrate models (e.g., Lakso et al., J. Neurochem., 86:165-72 (2003) and Link, Mech. Ageing Dev., 122:1639-49 (2001)). Exemplary molecular models of Type II diabetes include: a transgenic mouse having defective Nkx-2.2 or Nkx-6.1; (US 6,127,598); Zucker Diabetic Fatty fa/fa (ZDF) rat. (US 6569832); and Rhesus monkeys, which spontaneously develop obesity and subsequently frequently progress to overt type 2 diabetes (Hotta et al., Diabetes, 50:1126- 33 (2001); and a transgenic mouse with a dominant-negative IGF-I receptor (KR-IGF-IR) having Type 2 diabetes-like insulin resistance.
Exemplary animal and cellular models for neuropathy include: vincristine induced sensory-motor neuropathy in mice (US Appl 5420112) or rabbits (Ogawa et al., Neurotoxicology, 21:501-11 (2000)); a streptozotocin (STZ)-diabetic rat for study of autonomic neuropathy (Schmidt et al., Am. J. Pathol., 163:21-8 (2003)); and a progressive motor neuropathy (pmn) mouse (Martin et al., Genomics, 75:9-16 (2001)). With respect to neoplastic disorders, again, numerous animal and cellular models have been described. An exemplary in vivo system for evaluating a compound for its ability to limit the spread of primary tumors is described by Crowley et al., Proc. Natl. Acad. Sd., 90 : 5021-5025 (1993). Nude mice are injected with tumor cells (PC3) engineered to express CAT (chloramphenicol acetyltransferase). Compounds to be tested for their ability to decrease tumor size and/or metastases are administered to the animals, and subsequent measurements of tumor size and/or metastatic growths are made. The level of CAT detected in various organs provides an indication of the ability of the compound to inhibit metastasis; detection of less CAT in tissues of a treated animal versus a control animal indicates less CAT-expressing cells have migrated to that tissue or have propagated within that tissue.
Administration of compounds and formulations thereof
The compounds of the formulae described herein can, for example, be administered by injection, intravenously, intraarterially, subdermally, intraperitoneally, intramuscularly, or subcutaneously; or orally, buccally, nasally, transmucosally, topically, in an ophthalmic preparation, or by inhalation, with a dosage ranging from about 0.001 to about 100 mg/kg of body weight, e.g., between 0.001-lmg/kg, 1-lOOmg/kg, or 0.01- 5mg/kg, every 4 to 120 hours, e.g., about every 6, 8, 12, 24, 48, or 72 hours, or according to the requirements of the particular compound. The methods herein contemplate administration of an effective amount of compound or compound composition to achieve the desired or stated effect (e.g., reduction of feeding in a subject). Typically, the pharmaceutical compositions of this invention will be administered from about 1 to about 6 times per day, for example, the compounds can be administered about 1 to about 4 (e.g., 1, 2, 3, or 4) hours prior to meal time. Alternatively, the compounds can be administered as a continuous infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. A typical preparation will contain from about 5% to about 95% active compound (w/w). Alternatively, such preparations contain from about 20% to about 80% active compound.
Lower or higher doses than those recited above may be required. Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the disease, condition or symptoms, the patient's disposition to the disease, condition or symptoms, and the judgment of the treating physician.
Upon improvement of a patient's condition, a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
Pharmaceutical compositions of this invention comprise a compound of the formulae described herein or a pharmaceutically acceptable salt thereof; an additional compound including for example, a steroid or an analgesic; and any pharmaceutically acceptable carrier, adjuvant or vehicle. Alternate compositions of this invention comprise a compound of the formulae described herein or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier, adjuvant or vehicle. The compositions delineated herein include the compounds of the formulae delineated herein, as well as additional therapeutic compounds if present, in amounts effective for achieving a modulation of disease or disease symptoms, including kinase mediated disorders or symptoms thereof. The compositions are made by methods including the steps of combining one or more compounds delineated herein with one or more carriers and, optionally, one or more additional therapeutic compounds delineated herein.
The term "pharmaceutically acceptable carrier or adjuvant" refers to a carrier or adjuvant that may be administered to a patient, together with a compound of this invention, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the compound.
The pharmaceutical compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions and/or emulsions are administered orally, the active ingredient may be suspended or dissolved in an oily phase which can be combined with emulsifying and/or suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
The pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3- butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injec tables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms such as emulsions and or suspensions. Other commonly used surfactants such as Tweens or Spans and/or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
The pharmaceutical compositions of this invention may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-α-tocopherol polyethyleneglycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. Cyclodextrins such as α -, β-, and γ-cyclodextrin, may also be advantageously used to enhance delivery of compounds of the formulae described herein.
In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
When the compositions of this invention comprise a combination of a compound of the formulae described herein and one or more additional therapeutic or prophylactic agents, both the compound and the additional compound should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen. Additionally, combinations of a plurality of compounds described herein are also envisioned. The additional compounds may be administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those compounds may be part of a single dosage form, mixed together with the compounds of this invention in a single composition.
Treatments
The compounds described herein can be administered to cells in culture, e.g. in vitro or ex vivo, or to a subject, e.g., in vivo, to treat, prevent, and/or diagnose a variety of disorders, including those described herein below.
As used herein, the term "treat" or "treatment" is defined as the application or administration of a compound, alone or in combination with, a second compound to a subject, e.g., a patient, or application or administration of the compound to an isolated tissue or cell, e.g., cell line, from a subject, e.g., a patient, who has a disorder (e.g., a disorder as described herein), a symptom of a disorder, or a predisposition toward a disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disorder, one or more symptoms of the disorder or the predisposition toward the disorder (e.g., to prevent at least one symptom of the disorder or to delay onset of at least one symptom of the disorder).
As used herein, an amount of a compound effective to treat a disorder, or a "therapeutically effective amount" refers to an amount of the compound which is effective, upon single or multiple dose administration to a subject, in treating a cell, or in curing, alleviating, relieving or improving a subject with a disorder beyond that expected in the absence of such treatment.
As used herein, an amount of a compound effective to prevent a disorder, or "a prophylactically effective amount" of the compound refers to an amount effective, upon single- or multiple-dose administration to the subject, in preventing or delaying the occurrence of the onset or recurrence of a disorder or a symptom of the disorder.
As used herein, the term "subject" is intended to include human and non-human animals. Exemplary human subjects include a human patient having a disorder, e.g., a disorder described herein or a normal subject. The term "non-human animals" of the invention includes all vertebrates, e.g., non-mammals (such as chickens, amphibians, reptiles) and mammals, such as non-human primates, domesticated and/or agriculturally useful animals, e.g., sheep, dog, cat, cow, pig, etc.
Many compounds described herein can be used to treat or prevent a metabolic disorder. A "metabolic disorder" is a disease or disorder characterized by an abnormality or malfunction of metabolism. One category of metabolic disorders is disorders of glucose or insulin metabolism. For example, the subject can be insulin resistant, e.g., have insulin-resistance diabetes. In one embodiment, a compound described herein can be used to decrease insulin or glucose levels in a subject. In another embodiment, a compound described herein can be used to alter (e.g., increase) insulin or glucose levels in a subject. Treatment with a compound may be in an amount effective to improve one or more symptoms of the metabolic disorder.
In some instances, the invention provides a method of treating metabolic syndrome, including administering to a subject an effective amount of a compound described herein.
The metabolic syndrome (e.g., Syndrome X) is characterized by a group of metabolic risk factors in one person. They include: central obesity (excessive fat tissue in and around the abdomen), atherogenic dyslipidemia (blood fat disorders — mainly high triglycerides and low HDL cholesterol — that foster plaque buildups in artery walls); insulin resistance or glucose intolerance (the body can't properly use insulin or blood sugar); prothrombotic state (e.g., high fibrinogen or plasminogen activator inhibitor [-1] in the blood); raised blood pressure (i.e., hypertension) (130/85 mmHg or higher); and proinflammatory state (e.g., elevated high-sensitivity C-reactive protein in the blood).
The underlying causes of this syndrome are overweight/obesity, physical inactivity and genetic factors. People with metabolic syndrome are at increased risk of coronary heart disease, other diseases related to plaque buildups in artery walls (e.g., stroke and peripheral vascular disease) and type 2 diabetes. Metabolic syndrome is closely associated with a generalized metabolic disorder called insulin resistance, in which the body can't use insulin efficiently.
Many compounds described herein can be used to treat or prevent obesity, e.g., in a human subject, e.g. a child or adult subject. "Obesity" refers to a condition in which a subject has a body mass index of greater than or equal to 30. Many compounds described herein can be used to treat or prevent an over-weight condition. "Over-weight" refers to a condition in which a subject has a body mass index of greater or equal to 25.0. The body mass index (BMI) and other definitions are according to the "NIH Clinical Guidelines on the Identification and Evaluation, and Treatment of Overweight and Obesity in Adults" (1998). Treatment with the compound may be in an amount effective to alter the weight of the subject, e.g., by at least 2, 5, 7, 10, 12, 15, 20, 25, 30, 25, 40, 45, 50, or 55%. Treatment with the compound may be in an amount effective to reduce the body mass index of the subject, e.g., to less than 30, 28, 27, 25, 22, 20, or 18. The compounds can be used to treat or prevent aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., anorexia, bulimia, obesity, diabetes, or hyperlipidemia (e.g., elevated triglycerides and/or elevated cholesterol), as well as disorders of fat or lipid metabolism.
For example, agonists of GHS-R can be used to increase food intake or to treat disorders associated with weight loss, e.g., anorexia, bulimia, and so forth. Antagonists or inverse agonists of GHS-R can be used to treat aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., obesity, diabetes, or hyperlipidemia, as well as disorders of fat or lipid metabolism that results in weight gain. In one embodiment, a compound described herein is used to treat hypothalamic obesity. For example, the compound can be administered to a subject identified as at risk for hypothalamic obesity or to a subject that has an abnormal (e.g., extreme) insulin response to glucose.
In another embodiment, a compound described herein (e.g., a GHS-R antagonist or inverse agonist) can be administered to treat obesity associated with Prader-Willi Syndrome (PWS). PWS is a genetic disorder associated with obesity (e.g., morbid obesity). In general, individuals suffering from PWS also have deficient GH secretion. As opposed to individuals having common obesity, those individuals having PWS associated obesity have high fasting-ghrelin concentrations, which might contribute to hyperphagia. Accordingly, in some instances, a subject suffering from PWS associated obesity can be identified using genetic markers, determination of GH levels, fasting- ghrelin concentrations, careful phenotyping, or other methods known in the art.
Administration of a GHS-R antagonist such as one of the compounds described herein can be used to reduce body fat, prevent increased body fat, reduce cholesterol (e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol), and/or reduce appetite in individuals having PWS associated obesity, and/or reduce comorbidities such as diabetes, cardiovascular disease, and stroke.
In another embodiment, a compound described herein can be administered to treat fatty liver disease (FLD), e.g., non-alcoholic fatty liver disease. FLD, also known as steatorrhoeic hepatosis or steatosis hepatitis, is a reversible condition where large vacuoles of triglyceride fat accumulate in liver cells via the process of steatosis. The condition is also associated with other diseases that influence fat metabolism.
Many compounds described herein can be used to treat a neurological disorder. A "neurological disorder" is a disease or disorder characterized by an abnormality or malfunction of neuronal cells or neuronal support cells (e.g., glia or muscle). The disease or disorder can affect the central and/or peripheral nervous system. Exemplary neurological disorders include neuropathies, skeletal muscle atrophy, and neurodegenerative diseases, e.g., a neurodegenerative disease other than one caused at least in part by polyglutamine aggregation. Exemplary neurodegenerative diseases include: Alzheimer's, Amyotrophic Lateral Sclerosis (ALS), and Parkinson's disease. Another class of neurodegenerative diseases includes diseases caused at least in part by aggregation of poly-glutamine. Diseases of this class include: Huntington's Diseases, Spinalbulbar Muscular Atrophy (SBMA or Kennedy's Disease) Dentatorubropallidoluysian Atrophy (DRPLA), Spinocerebellar Ataxia 1 (SCAl), Spinocerebellar Ataxia 2 (SCA2), Machado-Joseph Disease (MJD; SCA3), Spinocerebellar Ataxia 6 (SCA6), Spinocerebellar Ataxia 7 (SCA7), and Spinocerebellar Ataxia 12 (SCA12). Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neurological disorder. In one embodiment, a compound having GHS-R antagonist activity can be used to treat the neurological disorder.
Many compounds described herein can be used to modulate anxiety in a subject. In one embodiment, a compound having, for example, GHS-R antagonist or inverse agonist activity can be used to decrease anxiety.
Many compounds described herein can be used to modulate memory retention in a subject. In one embodiment, a compound having GHS-R antagonist or inverse agonist activity can be used to decrease memory retention. For example, decreasing memory retention may aid recovery from traumatic stress. In one embodiment, a compound having GHS-R agonist activity is used to increase memory retention.
Many compounds described herein can be used to modulate sleep, sleep cycles (e.g., REM sleep), or wakefulness in a subject. In one embodiment, a compound having GHS-R agonist activity is used to promote sleep in the subject or to treat sleep apnea. In one embodiment, a GHS-R agonist, inverse agonist or antagonist (e.g., a compound described herein, is used to alter the circadian rhythm of a subject. For example, the compound can be delivered at particular times of day, e.g., regularly, e.g., in the evening and/or morning, to reset a circadian rhythm, e.g., prior to, during, or after traveling between time zones, or to a subject having a circadian disorder. The compounds can, e.g., modulate the pulsatility of GH secretion.
Many compounds described herein can be used to treat a cardiovascular disorder. A "cardiovascular disorder" is a disease or disorder characterized by an abnormality or malfunction of the cardiovascular system, e.g., heart, lung, or blood vessels. Exemplary cardiovascular disorders include: cardiac dysrhythmias, chronic congestive heart failure, ischemic stroke, coronary artery disease and cardiomyopathy. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the cardiovascular disorder, e.g., elevated triglyceride levels or elevated cholesterol. In one embodiment, a compound having GHS-R antagonist or inverse agonist activity can be used to treat the cardiovascular disorder.
Many compounds described herein can be used to treat a dermatological disorder or a dermatological tissue condition. A "dermatological disorder" is a disease or disorder characterized by an abnormality or malfunction of the skin. A "dermatological tissue condition" refers to the skin and any underlying tissue (e.g., support tissue), which contributes to the skin's function and/or appearance, e.g., cosmetic appearance. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the dermatological disorder or the dermatological tissue condition. In one embodiment, a compound having GHS-R antagonist or inverse agonist activity can be used to treat the dermatological disorder or dermatological tissue condition.
Many compounds described herein can be used to treat a geriatric disorder. A "geriatric disorder" is a disease or disorder whose incidence, at the time of filing of this application and in a selected population of greater than 100,000 individuals, is at least 70% among human individuals that are greater than 70 years of age. In one embodiment, the geriatric disorder is a disorder other than cancer or a cardio-pulmonary disorder. A preferred population is a United States population. A population can be restricted by gender and/or ethnicity.
Many compounds described herein can be used to treat or prevent a disorder characterized by excessive growth hormone activity. For example, the compounds can be used to reduce GH levels in the subject. In one embodiment, the subject is a human, e.g., a child (e.g., between 3-11 years), an adolescent (e.g., between 12-19 years), a young adult (e.g., between 20-25 years), or an adult. In one embodiment, a compound having GHS-R antagonist or inverse agonist activity is used to treat the disorder characterized by excessive growth hormone activity.
Many compounds described herein can be used to modulate vagal tone. For example, a compound described herein or other modulator of GHS-R can be administered to a subject who has a vagotomy or other disorder, which alters vagal afferent or efferent activity. In one embodiment, a subject is monitored for abnormalities in vagal nerve function, and, if a malfunction is detected, the subject is treated with a compound described herein or other modulator of GHS-R.
Exemplary diseases and disorders that are relevant to certain implementations include: cancer (e.g., breast cancer, colorectal cancer, CCL, CML, prostate cancer); skeletal muscle atrophy; adult-onset diabetes; diabetic nephropathy, neuropathy (e.g., sensory neuropathy, autonomic neuropathy, motor neuropathy, retinopathy); obesity; bone resorption; neurodegenerative disorders (Parkinson's disease, ALS, Alzheimer's, short- term and long-term memory loss) and disorders associated with protein aggregation (e.g., other than polyglutamine aggregation) or protein misfolding; age-related macular degeneration, Bell's Palsy; cardiovascular disorders (e.g., atherosclerosis, cardiac dysrhythmias, chronic congestive heart failure, ischemic stroke, coronary artery disease and cardiomyopathy), chronic renal failure, type 2 diabetes, ulceration, cataract, presbiopia, glomerulonephritis, Guillan-Barre syndrome, hemorrhagic stroke, rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, SLE, Crohn's disease, osteoarthritis, pneumonia, and urinary incontinence. Symptoms and diagnosis of diseases are well known to medical practitioners.
In certain embodiments, the compounds are directed locally to GHS-R in a target tissue of the organism. GHS-R is expressed in the hypothalamus, heart, lung, pancreas, intestine, brain (particularly in the arcuate nucleus (ARC)), and adipose tissue. A compound described herein can be targeted to one or more of the above tissues. For example, the compound can be formulated for inhalation for targeting to the lung. The compound can be formulated for ingestion, and passage to the intestine for targeting to the intestine. In other embodiments, treatment is directed systemically, and the compound is distributed to the target tissue.
Depending on the disorder and the compound, treatment may involve, in addition, to use of a compound in a class specified above, using a compound in another class. For example, in subjects whose endogenous ghrelin levels are lower than normal generally or lower than normal in an affected region, a treatment may involve using a compound having GHS-R agonist activity. In other subjects whose endogenous ghrelin levels are higher than normal generally or higher than normal in an affected region, treatment may involve using a compound having GHS-R antagonist activity. The suitability of a particular compound can be evaluated, e.g., in an animal -based assay or by monitoring a subject.
Many compounds described herein can be used to modulate activity of a biological signal that controls energy balance. Such signals include peptide signals, such as NPY, AGRP, orexins, MCH, beacon (see, e.g., Collier et al. (2000) Diabetes 49:1766), mealoncyte-stimulating hormone, neuromedin U, corticotrophin-releasing factor, and leptin. For example, NPY is a 36-amino acid peptide that stimulates food intact and depresses metabolic rate. Many compounds described herein can be used to decrease NPY activity. Many compounds described herein can be used to increase activity or availability of an anorexigenic molecule, e.g., bombesin, IL-lβ, leptin, and gastrin- releasing peptide. Accordingly, the compounds may increase the discharge rate of the gastric vagal afferent.
GHS-R expression in human tissues is generally expressed in pituitary cells, brain, spinal cord, uterus, spleen, pancreas, kidney, adrenal gland, skeletal muscle, thyroid, liver, small intestine, and heart. Accordingly, compounds described herein can be used to treat diseases and disorders associated with undesired levels of ghrelin or ghrelin-mediated signaling activity in those tissues. For example, if the level of ghrelin or ghrelin-mediated signaling activity is undesirably low, a compound having GHS-R agonist activity can be used for treatment. If the level of ghrelin or ghrelin-mediated signaling activity is undesirably high, a compound having GHS-R antagonist activity can be used for treatment. For example, the level of desired ghrelin activity can vary from tissue to tissue. Ghrelin is secreted by the stomach and may be high in or near the stomach, but much lower in normal pancreatic tissue.
Neoplastic Disorders
Many compounds described herein can be used to treat a neoplastic disorder. A "neoplastic disorder" is a disease or disorder characterized by cells that have the capacity for autonomous growth or replication, e.g., an abnormal state or condition characterized by proliferative cell growth. Exemplary neoplastic disorders include: carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors. Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
Whether a neoplastic disorder should be treated with a GHS-R agonist or antagonist can depend on the type of neoplasia. For example, Duxbury et al. (2003) Biochem. Biophys. Res. Comm. 309:464-468 report that certain neoplastic disorders are inhibited by GHS-R antagonists. These disorders include, e.g., pancreatic adenocarcinoma, and neoplasias in which GHS-R or GHS-RIb is expressed, e.g., prostate adenocarcinoma, pancreatic endocrine tumors, somatotroph tumors, and central nervous system tumors. Neoplasias that are attenuated, inhibited, or killed by a GHS-R antagonist are term, herein, "GHS-R antagonist -sensitive neoplastic disorders" and can be treated with a compound having GHS-R antagonist activity.
Duxbury et al. also report that certain other types of neoplasia, e.g., breast, lung, and thyroid adenocarcinomas can be inhibited by high levels ghrelin (> 10 nM) and, accordingly, can be treated with a GHS-R agonist, e.g., a GHS-R agonist described herein or another known GHS-R agonist. Neoplasias that are attenuated, inhibited, or killed by ghrelin or a GHS-R agonist are term, herein, "ghrelin-sensitive neoplastic disorders" and can be treated with a compound having GHS-R agonist activity.
Whether a neoplastic cell is sensitive to a ghrelin agonist or antagonist (i.e., whether the neoplastic cell is a ghrelin-sensitive or GHS-R antagonist sensitive neoplastic disorder) can be determined by a proliferation assay in the presence of a GHS-R agonist, e.g., ghrelin, or antagonist, e.g., D-Lys-GHRP6. Duxbury et al. disclose an exemplary proliferation assay. In one such assay, cells are seeded into 96 well plates with about 10 cells per well. The cells are cultured for 3 days in medium, and then contacted with ghrelin or D-Lys-GHRP6, or a control medium. Cells are then evaluated using the MTT assay (3-(4,5-dimethylthiazolyl-2yl)-2,5-diphenyltetrazolium) (from Trevigen, Gaithersburg, MD) for viability. Other assays that can be performed are invasion and migration assays. The affect of a particular compound may also depend on concentration, which can also be varied in the assay.
In addition to the above-mentioned neoplastic disorders, compounds described herein can be used to treat other neoplasias and hyperplasias including "tumors," which may be benign, premalignant or malignant.
Further examples of cancerous disorders include, but are not limited to, solid tumors, soft tissue tumors, and metastatic lesions. Examples of solid tumors include malignancies, e.g., sarcomas, adenocarcinomas, and carcinomas, of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract (e.g., renal, urothelial cells), pharynx, prostate, ovary as well as adenocarcinomas which include malignancies such as most colon cancers, rectal cancer, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, cancer of the small intestine and so forth. Metastatic lesions of the aforementioned cancers can also be treated or prevented using the methods and compositions of the invention.
A compound described herein can be useful in treating malignancies of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract, prostate, ovary, pharynx, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus. Exemplary solid tumors that can be treated include: fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, and retinoblastoma.
The term "carcinoma" is recognized by those skilled in the art and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas. Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary. The term also includes carcinosarcomas, e.g., which include malignant tumors composed of carcinomatous and sarcomatous tissues. An "adenocarcinoma" refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.
The term "sarcoma" is recognized by those skilled in the art and refers to malignant tumors of mesenchymal derivation.
The subject method can also be used to inhibit the proliferation of hyperplastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof. For instance, the invention contemplates the treatment of various myeloid disorders including, but not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Vaickus, L. (1991) Crit Rev. in Oncol./Hemotol. 11 :267-97). Lymphoid malignancies, which may be treated by the subject method, include, but are not limited to acute lymphoblastic leukemia (ALL), which includes B -lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and Waldenstrom's macroglobulinemia (WM). Additional forms of malignant lymphomas contemplated by the treatment method of the invention include, but are not limited to, non-Hodgkin's lymphoma and variants thereof, peripheral T-cell lymphomas, adult T-cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF) and Hodgkin's disease.
Modulators of GHS-R Compounds that modulate the activity of GHS-R can be used to treat a disorder in which a subject has less than a desired or less than a normal level of GHS-R activity, e.g., in a particular tissue; or more than a desired or higher than a normal level of GHS-R activity, e.g., in a particular tissue.
Many compounds described herein can be used to treat or prevent a metabolic disorder, e.g., glucose or insulin metabolism. For example, the subject can be insulin resistant, e.g., have insulin-resistance diabetes. In one embodiment, a compound described herein can be used to decrease insulin or glucose levels in a subject. In another embodiment, a compound described herein can be used to alter (e.g., increase) insulin or glucose levels in a subject.
In some instances, a compound described herein may be used to treat metabolic syndrome (e.g., Syndrome X), characterized by a group of metabolic risk factors in one person. They include: central obesity (excessive fat tissue in and around the abdomen), atherogenic dyslipidemia (blood fat disorders — mainly high triglycerides and low HDL cholesterol — that foster plaque buildups in artery walls); insulin resistance or glucose intolerance (the body can't properly use insulin or blood sugar); prothrombotic state (e.g., high fibrinogen or plasminogen activator inhibitor [-1] in the blood); raised blood pressure (i.e., hypertension) (130/85 mmHg or higher); and proinflammatory state (e.g., elevated high-sensitivity C-reactive protein in the blood). The underlying causes of this syndrome are overweight/obesity, physical inactivity and genetic factors. People with metabolic syndrome are at increased risk of coronary heart disease, other diseases related to plaque buildups in artery walls (e.g., stroke and peripheral vascular disease) and type 2 diabetes. Metabolic syndrome is closely associated with a generalized metabolic disorder called insulin resistance, in which the body can't use insulin efficiently.
Many compounds described herein can be used to treat or prevent obesity. The compounds can be used to treat or prevent aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., anorexia, bulimia, obesity, diabetes, or hyperlipidemia (e.g., elevated triglycerides and/or elevated cholesterol), as well as disorders of fat or lipid metabolism.
Many compounds described herein can be used to modulate anxiety in a subject. Many compounds described herein can be used to modulate sleep, sleep cycles (e.g., REM sleep), or wakefulness in a subject.
Many compounds described herein can be used to treat a cardiovascular disorder. Exemplary cardiovascular disorders include: cardiac dysrhythmias, chronic congestive heart failure, ischemic stroke, coronary artery disease and cardiomyopathy. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the cardiovascular disorder, e.g., elevated triglyceride levels or elevated cholesterol.
Many compounds described herein can be used to treat a geriatric disorder. In one embodiment, the geriatric disorder is a disorder other than cancer or a cardiopulmonary disorder.
Many compounds described herein can be used to modulate vagal tone. For example, a compound described herein or other modulator of GHS-R can be administered to a subject who has a vagotomy or other disorder, which alters vagal afferent or efferent activity. In one embodiment, a subject is monitored for abnormalities in vagal nerve function, and, if a malfunction is detected, the subject is treated with a compound described herein or other modulator of GHS-R.
Exemplary diseases and disorders that are relevant to certain implementations include: cancer (e.g., breast cancer, colorectal cancer, CCL, CML, prostate cancer); skeletal muscle atrophy; adult-onset diabetes; diabetic nephropathy, neuropathy (e.g., sensory neuropathy, autonomic neuropathy, motor neuropathy, retinopathy); obesity; bone resorption; neurodegenerative disorders (Parkinson's disease, ALS, Alzheimer's, short-term and long-term memory loss) and disorders associated with protein aggregation (e.g., other than polyglutamine aggregation) or protein misfolding; age-related macular degeneration, Bell's Palsy; cardiovascular disorders (e.g., atherosclerosis, cardiac dysrhythmias, chronic congestive heart failure, ischemic stroke, coronary artery disease and cardiomyopathy), chronic renal failure, type 2 diabetes, ulceration, cataract, presbiopia, glomerulonephritis, Guillan-Barre syndrome, hemorrhagic stroke, rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, SLE, Crohn's disease, osteoarthritis, pneumonia, and urinary incontinence. Symptoms and diagnosis of diseases are well known to medical practitioners. Many compounds described herein can be used to treat a neoplastic disorder, e.g., carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors. Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
Whether a neoplastic disorder should be treated with a GHS-R agonist or antagonist can depend on the type of neoplasia. For example, Duxbury et al. (2003) Biochem. Biophys. Res. Comm. 309:464-468 report that certain neoplastic disorders are inhibited by GHS-R antagonists. These disorders include, e.g., pancreatic adenocarcinoma, and neoplasias in which GHS-R or GHS-RIb is expressed, e.g., prostate adenocarcinoma, pancreatic endocrine tumors, somatotroph tumors, and central nervous system tumors.
Duxbury et al. also report that certain other types of neoplasia, e.g., breast, lung, and thyroid adenocarcinomas can be inhibited by high levels ghrelin (> 10 nM) and, accordingly, can be treated with a GHS-R agonist, e.g., a GHS-R agonist described herein or another known GHS-R agonist.
In addition to the above-mentioned neoplastic disorders, compounds described herein can be used to treat other neoplasias and hyperplasias including tumors, which may be benign, premalignant or malignant.
Further examples of cancerous disorders include, but are not limited to, solid tumors, soft tissue tumors, and metastatic lesions. Examples of solid tumors include malignancies, e.g., sarcomas, adenocarcinomas, and carcinomas, of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract (e.g., renal, urothelial cells), pharynx, prostate, ovary as well as adenocarcinomas which include malignancies such as most colon cancers, rectal cancer, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, cancer of the small intestine and so forth. Metastatic lesions of the aforementioned cancers can also be treated or prevented using the methods and compositions of the invention. A compound described herein can be useful in treating malignancies of the various organ systems, such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract, prostate, ovary, pharynx, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus. Exemplary solid tumors that can be treated include: fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, and retinoblastoma.
The subject method can also be used to inhibit the proliferation of hyperplastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof. For instance, the invention contemplates the treatment of various myeloid disorders including, but not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Vaickus, L. (1991) Crit Rev. in Oncol./Hemotol. 11 :267-97). Lymphoid malignancies, which may be treated by the subject method, include, but are not limited to acute lymphoblastic leukemia (ALL), which includes B -lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and Waldenstrom's macroglobulinemia (WM). Additional forms of malignant lymphomas contemplated by the treatment method of the invention include, but are not limited to, non-Hodgkin's lymphoma and variants thereof, peripheral T-cell lymphomas, adult T-cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF) and Hodgkin's disease.
Agonizing GHS-R
Compounds that agonize GHS-R can be used to treat a disorder in which a subject has less than a desired or less than a normal level of GHS-R activity, e.g., in a particular tissue. Such compounds can be used to for one of the following purposes: to increase food intake or to treat disorders associated with weight loss, e.g., anorexia, bulimia, etc.; to increase memory retention; to promote sleep or to treat sleep apnea; treating a neoplastic disorder, e.g., e.g., breast, lung, and thyroid adenocarcinomas that may be inhibited by high levels ghrelin; treating cachexia, wasting, stimulating growth hormone release in elderly humans, patients with cancer, heart failure, or AIDS; treating growth hormone deficient adults; prevention of catabolic side effects of glucocorticoids; treatment of osteoporosis; stimulation of the immune system, acceleration of wound healing; accelerating bone fracture repair; treatment of growth retardation; treating acute or chronic renal failure or insufficiency; treatment of physiological short stature, including growth hormone deficient children; treating short stature associated with chronic illness; treating obesity and growth retardation associated with obesity; treating growth retardation associated with Prader-Willi syndrome and Turner's syndrome; accelerating the recovery and reducing hospitalization of burn patients or following major surgery such as gastrointestinal surgery; treatment of intrauterine growth retardation, and skeletal dysplasia; treatment of hypercortisonism and Cushing's syndrome; treatment of peripheral neuropathies; treatment of osteochondrody-splasias, Noonans syndrome, sleep disorders, schizophrenia, depression, Alzheimer's disease, delayed wound healing, and psychosocial deprivation; treatment of pulmonary dysfunction and ventilator dependency; prevention or treatment of congestive heart failure, improving pulmonary function, restoring systolic and diastolic function, increasing myocardial contractility, decreasing peripheral total vascular resistance, diminishing or preventing loss of body weight and enhancing recovery following congestive heart failure; increasing appetite; attenuation of protein catabolic response after a major operation; treating malabsorption syndromes; reducing protein loss due to chronic illness such as cancer or AIDS; accelerating weight gain and protein accretion in patients on TPN (total parenteral nutrition); treatment of hyperinsulinemia; treatment of gastric and duodenal ulcers; stimulation of thymic development; adjunctive therapy for patients on chronic hemodialysis; treatment of immunosuppressed patients; enhancement of an antibody response, e.g., following vaccination; increasing the total lymphocyte count of a human; treatment of syndromes manifested by non-restorative sleep and musculoskeletal pain, including fibromyalgia syndrome or chronic fatigue syndrome; improvement in muscle strength, mobility, maintenance of skin thickness, metabolic homeostasis, renal hemeostasis in the frail elderly; stimulation of osteoblasts, bone remodelling, and cartilage growth; prevention and treatment of congestive heart failure; protection of cardiac structure and/or cardiac function; enhancing of recovery of a mammal following congestive heart failure; enhancing and/or improving sleep quality as well as the prevention and treatment of sleep disturbances; enhancing or improving sleep quality by increasing sleep efficiency and augmenting sleep maintenance; prevention and treatment of mood disorders, in particular depression; improving mood and subjective well being in a subject suffering from depression; reducing insulin resistance; stimulation of the immune system; stimulating and promoting gastric motility in patients after surgery or in gastroparesis secondary to degenerative conditions such as type II diabetes; and increasing growth. The compounds can be used to treat a human or an animal, e.g., livestock, a pet, etc.
Antagonizing GHS-R
Compounds that antagonize GHS-R can be used to treat a disorder in which a subject has higher than desired or higher than a normal level of GHS-R activity, e.g., in a particular tissue.
Antagonists or inverse agonists of GHS-R can be used to treat aberrant or inappropriate weight gain, metabolic rate, or fat deposition, e.g., obesity, diabetes, or hyperlipidemia, as well as disorders of fat or lipid metabolism that results in weight gain, hypothalamic obesity. For example, the compound can be administered to a subject identified as at risk for hypothalamic obesity or to a subject that has an abnormal (e.g., extreme) insulin response to glucose.
A compound described herein (e.g., a GHS-R antagonist or inverse agonist) can be administered to treat obesity associated with Prader-Willi Syndrome (PWS). Administration of a GHS-R antagonist such as one of the compounds described herein can be used to reduce body fat, prevent increased body fat, reduce cholesterol (e.g., total cholesterol and/or ratios of total cholesterol to HDL cholesterol), and/or reduce appetite in individuals having PWS associated obesity, and/or reduce comorbidities such as diabetes, cardiovascular disease, and stroke.
Many compounds described herein can be used to treat a neurological disorder. Exemplary neurological disorders include neuropathies, skeletal muscle atrophy, and neurodegenerative diseases, e.g., a neurodegenerative disease other than one caused at least in part by polyglutamine aggregation. Exemplary neurodegenerative diseases include: Alzheimer's, Amyotrophic Lateral Sclerosis (ALS), and Parkinson's disease. Another class of neurodegenerative diseases includes diseases caused at least in part by aggregation of poly-glutamine. Diseases of this class include: Huntington's Diseases, Spinalbulbar Muscular Atrophy (SBMA or Kennedy's Disease) Dentatorubropallidoluysian Atrophy (DRPLA), Spinocerebellar Ataxia 1 (SCAl), Spinocerebellar Ataxia 2 (SCA2), Machado-Joseph Disease (MJD; SCA3), Spinocerebellar Ataxia 6 (SCA6), Spinocerebellar Ataxia 7 (SCA7), and Spinocerebellar Ataxia 12 (SCA12).
A compound having GHS-R antagonist or inverse agonist activity may be used to treat anxiety.
A compound having GHS-R antagonist or inverse agonist activity can be used to decrease memory retention. For example, decreasing memory retention may aid recovery from traumatic stress.
AGHS-R agonist, inverse agonist or antagonist (e.g., a compound described herein) may used to alter the circadian rhythm of a subject. For example, the compound can be delivered at particular times of day, e.g., regularly, e.g., in the evening and/or morning, to reset a circadian rhythm, e.g., prior to, during, or after traveling between time zones, or to a subject having a circadian disorder. The compounds can, e.g., modulate the pulsatility of GH secretion.
In one embodiment, a compound having GHS-R antagonist or inverse agonist activity can be used to treat a cardiovascular disorder.
A compound having GHS-R antagonist or inverse agonist activity can be used to treat fatty liver disease (e.g., non-alcoholic fatty liver disease).
Many compounds described herein can be used to treat a dermatological disorder or a dermatological tissue condition. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the dermatological disorder or the dermatological tissue condition. In one embodiment, a compound having GHS-R antagonist or inverse agonist activity can be used to treat the dermatological disorder or dermatological tissue condition.
Many compounds described herein can be used to treat or prevent a disorder characterized by excessive growth hormone activity. For example, the compounds can be used to reduce GH levels in the subject. In one embodiment, the subject is a human, e.g., a child (e.g., between 3-11 years), an adolescent (e.g., between 12-19 years), a young adult (e.g., between 20-25 years), or an adult. In one embodiment, a compound having GHS-R antagonist or inverse agonist activity is used to treat the disorder characterized by excessive growth hormone activity.
Many compounds described herein can be used to treat a neoplastic disorder, e.g., carcinoma, sarcoma, metastatic disorders (e.g., tumors arising from prostate, colon, lung, breast and liver origin), hematopoietic neoplastic disorders, e.g., leukemias, metastatic tumors. Prevalent cancers include: breast, prostate, colon, lung, liver, and pancreatic cancers. Treatment with the compound may be in an amount effective to ameliorate at least one symptom of the neoplastic disorder, e.g., reduced cell proliferation, reduced tumor mass, etc.
Duxbury et al. (2003) Biochem. Biophys. Res. Comm. 309:464-468 report that certain neoplastic disorders are inhibited by GHS-R antagonists. These disorders include, e.g., pancreatic adenocarcinoma, and neoplasias in which GHS-R or GHS-RIb is expressed, e.g., prostate adenocarcinoma, pancreatic endocrine tumors, somatotroph tumors, and central nervous system tumors. A compound described herein may also be used to treat obesity (including inducing weight loss, reducing bodyweight, reducing food intake, reducing appetite, increasing metabolic rate, reducing fat intake, reducing carbohydrate craving, or inducing satiety); diabetes mellitus (including enhancing glucose tolerance and/or decreasing insulin resistance); type ii diabetes; hypertension; hyp erlipidcmia; osteoarthritis; cancer; gall bladder disease; sleep apnea: depression; compulsion; neuroses; insomnia/sleep disorder: substance abuse; pain: male and female sexual dysfunction (including male impotence, loss of libido, female sexual arousal dysfunction, female orgasmic dysfunction, hypoaeiive sexual desire disorder, sexual pain disorder and male erectile dysfunction); fever; inflammation; immune modulation; rheumatoid arthritis; and obesity related disorders.
The obesity-related disorders herein are associated with, caused by, or result from obesity. F.xamples of obesity-related disorders include overeating, binge eating, and bulimia, hypertension, diabetes, elevated plasma insulin concentrations and insulin resistance, dysiipidemias, hyperlipidemia, endometrial, breast, prostate and colon cancer, osteoarthritis, obstructive sleep apnea, cholelithiasis, gallstones, heart disease, abnormal heart rhythms and arrythmias, myocardial infarction, congestive heart failure, coronary heart disease, sudden death, stroke, polycystic ovary disease, craniopharyngioma, Prader- Willi Syndrome, ProhJieh's syndrome, ( ill-deficient subjects, normal variant short stature, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass, e.g, children with acute lymphoblastic leukemia. Further examples of obesity-related disorders are metabolic syndrome, insulin resistance syndrome, sexual and reproductive dysfunction, such as infertility, hypogonadism in males and hirsutism in females, gastrointestinal motility disorders, such as obesity-related gastroesophageal reflux, respiratory disorders, such as ohesity-hypoventilation syndrome (Pickwickian syndrome), cardiovascular disorders, inflammation, such as systemic inflammation of the vasculature, arteriosclerosis, hypercholesterolemia, hypemrieaerπia, lower back pain, gallbladder disease, gout, and kidney cancer, nicotine addiction, substance addiction and alcoholism. Obesity -induced or obesity -related co -morbidities include, but are not limited to, diabetes, non-insulin dependent diabetes mcliitus ■ type ii (2). impaired glucose tolerance, impaired fasting glucose, insulin resistance syndrome, dyslipidcmia, hypertension, hyperarieaeidcniia, gout, cardiovascular disease, stroke, coronary artery disease, myocardial infarction, angina pectoris, sleep apnea syndrome, Pickwickian syndrome, fatty liver, cerebral infarction, cerebral thrombosis, transient ischemic attack, orthopedic disorders, arthritis deformans, kimbodynia, emmcniopathy, and infertility, hi particular, co-morbidities include: hypertension, hyperlipidemia, dyslipidemia, glucose cardiovascular disease, sleep apnea, diabetes meilitus. and other obesity-related conditions.
Kits
A compound described herein can be provided in a kit. The kit includes (a) a composition that includes a compound described herein, and, optionally (b) informational material. The informational material can be descriptive, instructional, marketing or other material that relates to the methods described herein and/or the use of the compound described herein for the methods described herein.
The informational material of the kits is not limited in its form. In one embodiment, the informational material can include information about production of the compound, molecular weight of the compound, concentration, date of expiration, batch or production site information, and so forth. In one embodiment, the informational material relates to use of the compound described herein to treat a disorder described herein.
In one embodiment, the informational material can include instructions to administer the compound described herein in a suitable manner to perform the methods described herein, e.g., in a suitable dose, dosage form, or mode of administration (e.g., a dose, dosage form, or mode of administration described herein). Preferred doses, dosage forms, or modes of administration are parenteral, e.g., intravenous, intramuscular, subcutaneous, intraparenteral, bucosal, sublingual, intraoccular, and topical. In another embodiment, the informational material can include instructions to administer the compound described herein to a suitable subject, e.g., a human, e.g., a human having or at risk for a disorder described herein. For example, the material can include instructions to administer the compound described herein to such a subject.
The informational material of the kits is not limited in its form. In many cases, the informational material, e.g., instructions, is provided in printed matter, e.g., a printed text, drawing, and/or photograph, e.g., a label or printed sheet. However, the informational material can also be provided in other formats, such as computer readable material, video recording, or audio recording. In another embodiment, the informational material of the kit is contact information, e.g., a physical address, email address, website, or telephone number, where a user of the kit can obtain substantive information about a compound described herein and/or its use in the methods described herein. Of course, the informational material can also be provided in any combination of formats.
In addition to a compound described herein, the composition of the kit can include other ingredients, such as a solvent or buffer, a stabilizer, a preservative, and/or a second compound for treating a condition or disorder described herein. Alternatively, the other ingredients can be included in the kit, but in different compositions or containers than the compound described herein. In such embodiments, the kit can include instructions for admixing the compound described herein and the other ingredients, or for using a compound described herein together with the other ingredients.
The compound described herein can be provided in any form, e.g., liquid, dried or lyophilized form. It is preferred that the compound described herein be substantially pure and/or sterile. When the compound described herein is provided in a liquid solution, the liquid solution preferably is an aqueous solution, with a sterile aqueous solution being preferred. When the compound described herein is provided as a dried form, reconstitution generally is by the addition of a suitable solvent. The solvent, e.g., sterile water or buffer, can optionally be provided in the kit.
The kit can include one or more containers for the composition containing the compound described herein. In some embodiments, the kit contains separate containers, dividers or compartments for the composition and informational material. For example, the composition can be contained in a bottle, vial, or syringe, and the informational material can be contained in a plastic sleeve or packet. In other embodiments, the separate elements of the kit are contained within a single, undivided container. For example, the composition is contained in a bottle, vial or syringe that has attached thereto the informational material in the form of a label. In some embodiments, the kit includes a plurality (e.g., a pack) of individual containers, each containing one or more unit dosage forms (e.g., a dosage form described herein) of a compound described herein. For example, the kit includes a plurality of syringes, ampules, foil packets, or blister packs, each containing a single unit dose of a compound described herein. The containers of the kits can be air tight, waterproof (e.g., impermeable to changes in moisture or evaporation), and/or light-tight.
The kit optionally includes a device suitable for administration of the composition, e.g., a syringe, inhalant, pipette, forceps, measured spoon, dropper (e.g., eye dropper), swab (e.g., a cotton swab or wooden swab), or any such delivery device. In a preferred embodiment, the device is an implantable delivery device.
EXAMPLES
Example 1. Synthesis of Compound 5.
1 TFA 2 DPPA/Et3N/tBuOH
Figure imgf000057_0001
Figure imgf000057_0002
Preparation of t-butyl diketo ester 2:
To a solution of t-butyl acetate (4 eq in THF), 3.5 eq of 25% THF solution of lithium hexamethyldisilazide will be added slowly at -78°C through a canula and stirred for 30 minutes. To this, a solution of ester 1 (1 eq in THF) will be added slowly and continued to stir at -78°C for 2 hrs. The progress of the reaction will be monitored by TLC. The reaction mixture will be quenched with water, extracted with ethyl acetate and the combined ethyl acetate layer dried over Na2SO4 and concentrated to remove the solvent. The crude product will be purified on silica gel (60-120 mesh) to yield the diketo ester 2.
Preparation of isoxazole 3:
Diketo ester 2 (1 eq) will be dissolved in ethanol followed by addition of potassium carbonate (2.5 eq) at 00C and allowed to stir at room temperature for 20 minutes. After 20 minutes, 2,6-dichlorophenyl-N-oxide (1.2 eq) will be added and the reaction mixture will be refluxed for 2 hrs. The progress of the reaction will be monitored by TLC. The reaction mixture will be filtered through a Buchner funnel and the filtrate concentrated and purified by silica gel column chromatography (60-120 mesh) to yield isoxazole compound 3.
Preparation of isoxazole NHBoc 4:
Isoxazole ester 3 will be dissolved in trifluoroacetic acid and stirred at room temperature for 2 hrs. The progress of the reaction will monitored by TLC. The excess solvent will be removed under reduced pressure, and the product will be dried and washed with hexane to furnish the TFA salt of the isoxazole carboxylic acid. This product will be dissolved in toluene followed by the addition of triethylamine (4 eq) at 00C. To this diphenylphosphoryl azide (1.1 eq) will be added and continued to stir for 20 minutes after which t-butyl alcohol will be added to the reaction mixture and refluxed at 110 0C for another 4 hrs. Progress of the reaction will be monitored by TLC. Excess solvents will be removed under reduced pressure and the crude product will be purified using silica gel column chromatography (60-120 mesh) to yield isoxazole NHBoc compound 4.
Preparation of sulfonamide 5: Boc-protected compound 4 (1 eq) will be dissolved in saturated MeOH/HCl and allowed to stir at room temperature for 1 hr. The solvent will removed under reduced pressure and the resulting deprotected amine hydrochloride salt will be washed with ether and used as such for the next step without further purification. The product isoxazole amine (1.0 eq) will be suspended in DMF (40 ml) followed by the addition of triethylamine (4 eq) at 00C. To this, a solution of sulfonyl chloride (2.0 eq) will be added at the same temperature and allowed to stir at room temperature for 1 hr and 30 minutes. The progress of the reaction will be monitored by TLC. The reaction mixture will be quenched with saturated sodium bicarbonate solution and then extracted with ethyl acetate. The combined organic layer will be dried over Na2SO4, concentrated and the residue purified using silica gel column chromatography (60-120 mesh). After concentration of the column fractions, the final product will be diluted with DCM and washed with water to remove the polar impurities, concentrated and stirred with pentane to obtain the final sulfonamide 5.
Procedure for the preparation of 2,6-dichlorophenyl-N-oxide:
Figure imgf000059_0001
Preparation of N-oxime b:
2,6-dichlorobenzaldehyde (25 gm, 0.143 moles, 1 eq) was dissolved in EtOH (200 ml) followed by the addition of hydroxyl amine hydrochloride (19.9 gm, 0.286 moles, 2 eq) and pyridine (34 gm, 0.429 moles, 3 eq) at 00C. The reaction mixture was refluxed for 40 minutes and checked by TLC. The solvent was removed under reduced pressure and to the residue water was added and extracted with ethyl acetate twice. The combined organic layer was washed with brine, dried over Na2SO4, concentrated and washed with chilled hexane to afford the mixture of syn and anti oxime b in 85% yield (23 gm). MS 190.0 (M+ peak) 1H NMR (200 MHz, CDCl3): δ 7.20-7.30 (m, 3H), 7.38-4.00 (m, 3H), 8.20 (s, IH), 8.40 (s, IH) mixture of syn and anti isomers.
Preparation of N-oxide c:
Oxime b (23 gm, 0.122 moles, 1 eq) was dissolved in DCM (250 ml) and to this N-chlorosuccinimide (19.6 gm, 0.1464 moles, 1.2 eq) was added at 00C followed by the addition of pyridine (13.5 gm, 0.1708 moles, 1.4 eq). The reaction mixture was stirred at 00C for 30 minutes and monitored by TLC. The reaction mass was washed with H2O (500 ml) and the DCM layer was separated, dried over Na2SO4, concentrated and purified immediately by fast column (silica gel, 60-120 mesh, 3-5% EA/hexane). Pale yellow solid of N-oxide c was obtained in 61% yield (14 gm) and used immediately for the next reaction. All the concentrations were done below 45 0C and any increase in the temperature resulted in the decomposition of N-oxide.

Claims

WHAT IS CLAIMED IS:
1. A compound of formula (I),
Figure imgf000061_0001
formula (I) or a therapeutically suitable salt or prodrug thereof, wherein:
R1 is alkoxyalkyl, alkyl, alkylC(O)NHalkyl, alkylS(O)2NHalkyl, alkenyl, aryl, heteroaryl, cyclyl, heterocyclyl, hydroxyalkyl, RaRbN-, RaRbNalkyl-, RaRbNcarboxyalkyl-, or -(CR10R10 )mR5; wherein R1 is optionally independently substituted with 1, 2, or 3 R6; m is 0, 1, 2, 3, or 4;
R2 is a member selected from the group consisting of alkyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkenylalkyl, heterocycle and heterocyclealkyl, each of which is optionally substituted with -NR8R8 ;
R3 and R4 are each members independently selected from the group consisting of hydrogen, alkyl, alkoxy, aryl, halogen, haloalkyl, cycloalkyl, cyano and nitro;
R5 is cyano, nitro, hydroxy, oxo, -OR7, -C(O)R7, -C(O)OR7, -OC(O)R7, -NR8R8', - C(O)NR8R8', -NR8C(O)R7, -NR8C(O)NR8R8', -OC(O)NR8R8', -NR8C(O)OR7, - SC(O)NR8R8', -NR8C(O)SR7, -SR9, -NR8SO2R9, -SO2NR8R8', -SOR9, and -S(O)2R9; wherein R5 is optionally substituted with 1, 2, or 3 R6 as described for R1; each R6 is independently halo, alkyl, cyclyl, heterocyclyl, aryl, heteroaryl, haloalkyl, haloalkyloxy, haloalkylthio, acyl, cyano, nitro, hydroxy, alkoxy, hydroxyalkyl, alkoxyalkyl, oxo, amino, alkylamino, dialkylamino, thiol, alkylthiol, -alkylCOOalkyl, or -alkylCOOH, CONR10R10', -C(O)alkyl, -C(O)arylalkyl; R7 is alkyl, alkenyl, alkynyl, cyclyl, cyclylalkyl, heterocyclyl, heterocyclylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, or haloalkyl; wherein R7 may be optionally substituted as provided in R5; each R8 and R8 is independently hydrogen, acyl, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, cyclyl, or cyclylalkyl, or R8 and R8 taken together can be cyclized to form -(CH2)qX(CH2)sS wherein each R8 and R8 may independently optionally be substituted as provided in R ;
R9 is alkyl, alkenyl, alkynyl, cyclyl, cyclylalkyl, heterocyclyl, heterocyclylalkyl, aryl, arylalkyl heteroaryl, heteroarylalkyl, or haloalkyl; wherein R may be optionally substituted as provided in R5; q is O, 1, 2, 3 or 4; s is 0, 1, 2, 3, or 4;
X is CR10R10', O, S, S(O), S(O)2, or NR10; each R and R is independently hydrogen or alkyl;
Ra and R are each members independently selected from the group consisting of hydrogen, alkoxyalkyl, alkyl, alkylcarbonyl, alkylsulfonyl, aryloxyalkyl and
R0R Ncarboxyalkylcarbonyl; and
RRcc aanndd RRd aarree each independently a member selected from the group consisting of hydrogen, and alkyl.
2. The compound of claim 1, wherein R and R are hydrogen.
3. The compound of claim 1 , wherein R is dialkylaminoalkyl.
^i
4. The compound of claim 3, wherein R *2 i •s
5. The compound of claim 1, wherein R1 is -(CR10R10^)1nR5.
6. The compound of claim 5, wherein R10 and R10 are hydrogen.
7. The compound of claim 1, wherein m is 1.
8. The compound of claim 1, wherein m is 2.
9. The compound of claim 5, wherein R5 is NR8C(O)R7.
10. The compound of claim 9, wherein R is hydrogen.
11. The compound of claim 9, wherein R is alkyl.
12. The compound of claim 11, wherein R is methyl.
13. The compound of claim 9, wherein R is aryl.
14. The compound of claim 13, wherein R is phenyl.
15. The compound of claim 9, wherein R is heteroaryl.
16. The compound of claim 15, wherein R is thiazolyl.
17. The compound of claim 5, wherein R5 is -NR8C(O)OR7.
18. The compound of claim 17, wherein R is hydrogen.
19. The compound of claim 17, wherein R is alkyl.
20. The compound of claim 17, wherein R8 is methyl.
21. The compound of claim 17, wherein R7 is cyclyl.
22. The compound of claim 17, wherein R7 is cyclopropyl.
23. The compound of claim 5, wherein R5 is NR8C(O)NR8R8
24. The compound of claim 23, wherein R is hydrogen.
25. The compound of claim 23, wherein R and R taken together are cyclized to form -(CH2)C1X(CH2)S-.
26. The compound of claim 23, wherein R and R taken together are cyclized to form a piperidine ring.
27. The compound of claim 5, wherein R5 is OC(O)NR8R8
28. The compound of claim 27, wherein one of R or R is H.
29. The compound of claim 27, wherein neither R or R is H.
30. The compound of claim 27, wherein R is alkyl.
31. The compound of claim 30, wherein R is methyl.
32. The compound of claim 30, wherein R is isopropyl.
33. The compound of claim 27, wherein R is alkyl.
34. The compound of claim 33, wherein R is methyl.
35. A method of treating a disorder regulated by ghrelin including anorexia, cancer cachexia, eating disorders, age-related decline in body composition, weight gain, obesity, and diabetes mellitus, comprising administering to a subject a compound of claim 1, formula (I).
36. A pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1, formula (I), in combination with a pharmaceutically suitable carrier.
PCT/US2010/026849 2009-03-10 2010-03-10 Sulfonamide containing compounds and uses thereof WO2010104967A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US15897609P 2009-03-10 2009-03-10
US61/158,976 2009-03-10

Publications (1)

Publication Number Publication Date
WO2010104967A1 true WO2010104967A1 (en) 2010-09-16

Family

ID=42728758

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/026849 WO2010104967A1 (en) 2009-03-10 2010-03-10 Sulfonamide containing compounds and uses thereof

Country Status (1)

Country Link
WO (1) WO2010104967A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060089398A1 (en) * 2003-03-19 2006-04-27 Gang Liu Isoxazole carboxamide derivatives as ghrelin receptor modulators
WO2008008286A2 (en) * 2006-07-12 2008-01-17 Merck & Co., Inc. Substituted pyrazoles as ghrelin receptor antagonists

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060089398A1 (en) * 2003-03-19 2006-04-27 Gang Liu Isoxazole carboxamide derivatives as ghrelin receptor modulators
WO2008008286A2 (en) * 2006-07-12 2008-01-17 Merck & Co., Inc. Substituted pyrazoles as ghrelin receptor antagonists

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BECKNELL ET AL.: "Novel C-3 N-urea, amide, and carbamate dihydroindazolo[5,4-a]pyrrolo[3,4- c]carbazole analogs as potent TIE-2 and VEGF-R2 dual inhibitors.", BIOORG MED CHEM LETT., vol. 16, no. 20, 2006, pages 5368 - 72 *
ROARK ET AL.: "Inhibitors of Acyl-CoA:Cholesterol Acyltransferase (ACAT). 2. Modification of Fatty Acid Anilide ACAT Inhibitors: Bioisosteric Replacement of the Amide Bond.", J. MOD. CHEM., vol. 36, 1993, pages 1662 - 1668 *

Similar Documents

Publication Publication Date Title
US7829589B2 (en) Sulfonamide compounds and uses thereof
US7897765B2 (en) Therapeutic compounds and uses thereof
US20050261332A1 (en) Sulfonamides and uses thereof
US20120088747A1 (en) Sulfonamide containing compounds and uses thereof
WO2006135860A1 (en) Sulfonamide compounds and uses thereof
EP1637521B1 (en) Novel tricyclic heterocycle compound
US20080261970A1 (en) Sufonamides and Uses Thereof
EP1572113B1 (en) Calcium receptor modulating compound and use thereof
WO2008141189A1 (en) Ghrelin modulating compounds and combinations thereof
US11046660B2 (en) Compounds and their use as PDE4 activators
EP3271338B1 (en) Triazole derivatives and their use as pde4 activators
US20210032231A1 (en) Compounds and Their Use as PDE4 Activators
US20180118728A1 (en) Novel pyridinium compounds
WO2010104929A1 (en) Sulfonamide containing compounds and uses thereof
WO2010104967A1 (en) Sulfonamide containing compounds and uses thereof
JP2006516647A (en) Novel substituted 2,3-benzodiazepine derivatives

Legal Events

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

Ref document number: 10751369

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10751369

Country of ref document: EP

Kind code of ref document: A1