WO2008141189A1 - Ghrelin modulating compounds and combinations thereof - Google Patents

Ghrelin modulating compounds and combinations thereof Download PDF

Info

Publication number
WO2008141189A1
WO2008141189A1 PCT/US2008/063257 US2008063257W WO2008141189A1 WO 2008141189 A1 WO2008141189 A1 WO 2008141189A1 US 2008063257 W US2008063257 W US 2008063257W WO 2008141189 A1 WO2008141189 A1 WO 2008141189A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
agent
iii
administered
Prior art date
Application number
PCT/US2008/063257
Other languages
French (fr)
Inventor
Alan Watson
Peter Distefano
Bard J. Geesaman
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 WO2008141189A1 publication Critical patent/WO2008141189A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds

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.
  • compounds that modulate GHS-R activity are at least useful for controlling disorders associated with GHS-R physiology.
  • 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.
  • heteroaryl compounds include oxadiazole and triazole 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.
  • the compounds, including stereoisomers thereof, can be created either singly, in small clusters, or in a combinatorial fashion to give structurally diverse libraries of compounds.
  • the invention features a compound of formula (I)
  • R 2 is hydrogen, halo (e.g., fluoro), Ci-C 6 alkyl, C 2 -C ⁇ 5 alkenyl, or C 2 -C 6 alkynyl; or R 2 can be taken together with R 1 to form a ring;
  • R 3 is hydrogen, Ci-C ⁇ alkyl, C 2 -Cg alkenyl, or C 2 -Ce alkynyl, or R 3 can be taken together with R 2 , R 4 , or R 5 to form a ring; each of which can be optionally substituted with 1-2 R 6' ;
  • x and y are each independently 0-6;
  • M is aryl, heteroaryl, cyclyl, or heterocyclyl, each of which is optionally substituted with 1-4 R 9 ;
  • R 4 and R 3 arc each independently hydrogen, alkyl, alkenyl, haloalkyl, cyclyl, or heterocyclyl, or R 4 and R 1 can be taken together to form a heterocyclic ring, or R 4 and R 5 can be taken together to form an azido moiety, or one or both of R 4 and R 5 can independently be joined to one or both of R 7a and R 7b to form one or more bridges between the nitrogen to which the R 4 and R 5 are attached and R 7a and R 7b , wherein each bridge contains 1 to 5 carbons; or one or both of R 4 and R 5 can independently be joined to one or both of R 7a and R 7b to form to form one or more heterocyclic rings including the nitrogen to which the R 4 and R 5 are attached, or one or both of R 4 and R 5 can independently be joined to R 3 to form a
  • Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R 10 ; each R 6 and R 6 are independently halo, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl, heteroaryl, alkoxy, haloalkyl, haloalkyloxy, haloalkylthio, acetyl, cyano, nitro, hydroxy, oxo, C(O)OR 2 , OC(O)R 2 , N(R 3 ) 2 , C(O)N(R 3 ) 2 , NR 3 C(O)R 2 , or
  • R 7a and R 7b are each independently hydrogen, alkyl, alkenyl, haloalkyl, cyclyl, cyclylalkyl, or heterocyclyl; or one or both of R 7a and R 7b can independently be joined to one or both of R 4 and R 5 to form one or more bridges between the nitrogen to which the R 4 and R 5 are attached and R 7a and R 7b , wherein each bridge contains 1 to 5 carbons; or one or both of R 7a and R 7b can independently be joined to one or both of R 4 and R 5 to form to form one or more heterocyclic rings including the nitrogen to which the R 4 and R 5 are attached, or one or both of R 7a and R 7 * 5 can independently be joined with R 8 to form a ring; wherein each R 7a and R 7b can be independently optionally substituted with 1- 5 halo, 1-3 hydroxy, 1-3 alkyl, 1-3 alkoxy, 1-3 amino, 1-3 alkylamino, 1-3 dialklyamino,
  • R 8 is hydrogen or Ci-C 6 alkyl, or R 8 can be joined with R 4 , R 5 , R 7a or R 7b to form a ring;
  • R 9 is halo, alkyl, cyclyl, heterocyclyl, aryl, heteroaryl, alkoxy, haloalkyl, haloalkyloxy, haloalkylthio, acetyl, cyano, nitro, hydroxy, oxo, C(O)OR 2 , OC(O)R 2 , N(R 2 ) 2 , C(O)N(R 2 ) 2 , NR 2 C(O)R 2 , SR 2 ; each R 10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl,
  • R 11 and R 11' are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
  • R 12 and R 12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R 12 and R 12 taken together can be cyclized to form -(CH2) q X(CH 2 )s-; wherein each R 12 and R 12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR 11 , alkoxy, heterocycloalkyl, -NR 11 C(O)NR 11 R 11' , -C(O)NR 11 R 11' , -NR 11 C(O)R 11' , -CN, oxo, -NR 11 SO 2 R 11' , -OC(O)R 11 , -SO 2 NR 11 R 11' , -S
  • X is CR 11 R 11' , O, S, S(O), S(O) 2 , or NR 11 ;
  • m is an integer between 1 and 6;
  • p is an integer from O to 5;
  • q and s are each independently an integer between 1 and 3; and
  • w is an integer between 0 and 5.
  • formula (I) comprises an enriched preparation of formula
  • formula (I) comprises an enriched preparation of formula (I")
  • R 1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl. In some embodiments, n is 1; k' is O; and R 1 is arylalkyl.
  • R 1 can be phenylmethyl.
  • n is 2; k' is a bond; and R 1 is aryl.
  • R 1 can be a branched alkyl such as one of the following .
  • R 2 is hydrogen or C1-C3 alkyl.
  • n is 0 and k' is a bond.
  • Exemplary R 1 moieties include methyl, and ethyl.
  • Preferred R 1 moieties include methyl.
  • R 1 is unsubstituted methyl or methyl or ethyl substituted with C(O)N(R 3 )2.
  • n is 0 and k' is a bond, and R 1 and R 2 are both methyl.
  • R 1 is hydrogen
  • R 3 is hydrogen
  • R and R together form a heterocyclic ring such as a pyrrolidine or an azetidine ring.
  • the heterocyclic ring can be unsubstituted or substituted, for example, with 1-2 R 6 .
  • R 1 and R 2 together form a ring.
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A can be any organic compound having the same or a same shape.
  • A can be any organic compound having the same or a same shape.
  • A can be any organic compound having the same or a same shape.
  • A can be any organic compound having the same or a same shape.
  • R 7a and R 7b are H; x is 1 ; and y is 0 or 1. In some embodiments,
  • A is CH 2 CH 2 or CH 2 CH 2 CH 2 ; and each R 4 and R 5 is independently alkyl, or R 4 and R 5 , when taken together, form a heterocyclic ring.
  • R' a and R can each be H.
  • At least one of R 7a or R 7b is taken together with at least one of R 4 or R 5 to form a heterocyclic ring including the nitrogen to which the R 4 and R 5 are attached.
  • R 7a and R 7b are each independently alkyl; R 4 and R 5 are each independently hydrogen or alkyl; and x and y are each independently 0 or 1. In some embodiments,
  • Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
  • Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
  • Y is substituted with one R 10 .
  • R 10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon.
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroary alkyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety.
  • the cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • Y is oxadiazole or triazole.
  • the invention features a compound of formula (II),
  • the compound of formula (II) comprises an enriched preparation of formula (II')
  • the compound of formula (II) comprises an enriched preparation of formula (H")
  • Q 1 and Q 4 are each independently S, O, N, or NR 10 . In some embodiments, Q 1 and Q 3 are each independently S, O, N, or NR 10 .
  • Q 2 is CR 2 or CR 10 .
  • Q 2 is S, O, N, or NR 10 .
  • At least one of Q 2 or Q 3 is CR 2 or CR 10 .
  • At least two of Q 1 , Q 2 , Q 3 , or Q 4 is S, O, N, or NR 10 .
  • Q 1 , Q 2 , and Q 3 are each independently S, O, N, or NR 10 .
  • Q 1 is NR 10 .
  • one of Q 2 , Q 3 , or Q 4 is CR 2 '
  • Q 2 is CR 10 .
  • Q 3 is CR 2 ' In some embodiments, Q 1 , Q 2 , Q 3 and Q 4 together form
  • Q 1 is NR 2 .
  • Q , Q , Q and Q together form
  • Q 1 is NR 10 .
  • the invention features a compound of formula (III),
  • Z , Z , Z , Z , and Z together form an aryl or heteroaryl moiety, and each Z , Z ,
  • Z 3 , Z 4 , and Z 5 is independently N, CR 10 , or CR 2 .
  • the compound of formula (III) comprises an enriched preparation of formula (HF)
  • the compound of formula (III) comprises an enriched preparation of formula (HF)
  • one of Z 1 , Z 2 , Z 3 , Z 4 , and Z 5 is N.
  • two of Z 1 , Z 2 , Z 3 , Z 4 , and Z 5 are N.
  • three of Z 1 , Z 2 , Z 3 , Z 4 , and Z 5 is N.
  • two of Z 1 and Z 2 are N. In some embodiments, two of Z 1 and Z 3 are N.
  • two of Z 1 and Z 4 are N.
  • two of Z 1 , Z 3 , and Z 5 are N.
  • the compound is a compound of formula (I), wherein Y is substituted with a single substituent R 10 .
  • R 10 can be aryl or heteroaryl, optionally substituted with up to three independent R 16 .
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl. In some embodiments, R 10 is substituted with 1-3 R 16 . In some embodiments, R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety.
  • the cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is R ⁇ .
  • Y is substituted with a second R 10 , for example an alkyl, halo or alkoxy.
  • R 1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl; k' is a bond or O; n is 1 or 2;
  • R 2 and R 3 are both hydrogen
  • x and y are each independently 0-6; R 4 and R 5 are each independently hydrogen or alkyl;
  • Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R 10 ; each R 10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR 11 , - NR 11 R 11' , -CF 3 , -SOR 12 , -SO 2 R 12 , -OC(O)R 11 , -SO 2 NR 12 R 12' , -(CH 2 ) m R 14 or R 15 ; each of which is optionally independently substituted with 1-3 R 16 ; R 11 and R 11 are each independently
  • R 12 and R 12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R 12 and R 12 taken together can be cyclized to form -(CH 2 ) q X(CH 2 ) s -; wherein each R 12 and R 12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR 11 , alkoxy, heterocycloalkyl, -NR 11 C(O)NR 11 R 11' , -C(O)NR 11 R 11' , -NR 11 C(O)R 11' , -CN, oxo, -NR 11 SO 2 R 11' , -OC(O)R 11 , -SO 2 NR 11 R 11'
  • X is CR 11 R 11' , O, S, S(O), S(O) 2 , or NR 11 ;
  • m is an integer between 1 and 6;
  • p is an integer from O to 5;
  • q and s are each independently an integer between 1 and 3; and
  • w is an integer between O and 5.
  • n is 1; k' is a bond or O; and R 1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl.
  • n is 1 ; k' is O; and R 1 is arylalkyl, for example phenylmethyl.
  • n is 2; k'is a bond; and R 1 is aryl.
  • R 7a and R 7b are H; x is 1 ; and y is O or 1.
  • A is CH 2 CH 2 or CH 2 CH 2 CH 2 CH 2 .
  • each R 4 and R 5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
  • Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
  • Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
  • Y is substituted with one R 10 .
  • R 10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon.
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroary alkyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety.
  • the cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • Y is oxadiazole or triazole.
  • Y is "Q , wherein Ql is O or NR 2 , preferably O or NH.
  • R 10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R 16 .
  • R 10 is substituted with one R 16 , such as halo (e.g., fluoro or chloro) or alkoxy.
  • the compounds has a formula (Ia)
  • R 1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl; k' is a bond or O; n is 1 or 2; A is CH 2 , CH 2 CH 2 , or CH 2 CH 2 CH 2 ;
  • R 4 and R ⁇ are each independently hydrogen or alkyl
  • Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R 10 ; each R 10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR 11 , - NR 11 R 11' , -CF 3 , -SOR 12 , -SO 2 R 12 , -OC(O)R 11 , -SO 2 NR 12 R 12' , -(CH 2 ) m R 14 or R 15 ; each of which is optionally independently substituted with 1-3 R 16 ;
  • R 11 and R 11 are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
  • R 12 and R 12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R 12 and R 12 taken together can be cyclized to form -(CH 2 ) q X(CH 2 )s-; wherein each R 12 and R 12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR 11 , alkoxy, heterocycloalkyl, -NR 11 C(O)NR 11 R 11' , -C(O)NR 11 R 11' , -NR 11 C(O)R 11' , -CN, oxo, -NR 11 SO 2 R 11 ', -OC(O)R 11 , -SO 2 NR 11 R 11' ,
  • each R 15 is independently -(CH 2 ) P N(R 12 )C(O)R 12' , -(CH 2 ) P CN, - (CH 2 ) p N(R 12 )C(O)OR 12' , -(CH 2 ) P N(R 12 )C(O)NR 12 R 12' , -(CH 2 ) P N(R 12 )SO 2 R 12 , - (CH 2 ) P SO 2 NR 12 R 12' , -(CH 2 ) p C(O)NR 12 R 12' , -(CH 2 ) P C(O)OR 12 , -(CH 2 ) P OC(O)OR 12 , - (CH 2 )pOC(O)R 12 , - (CH 2 )pOC(O)R 12 , - (CH 2 )pOC(O)R 12 , - (CH 2 )pOC(O)R 12 , - (CH 2 )pOC(O
  • X is CR 11 R 11' , O, S, S(O), S(O) 2 , or NR 11 ;
  • m is an integer between 1 and 6;
  • p is an integer from O to 5;
  • q and s are each independently an integer between 1 and 3; and
  • w is an integer between O and 5.
  • n is 1; k' is a bond or O; and R 1 is aryl, heteroaryl, arylalkyl, or heteroarylatkyl.
  • n is 1; k' is O; and R 1 is arylalkyl, for example phenylmethyl.
  • n is 2; k'is a bond; and R 1 is aryl.
  • A is CH 2 CH 2 or CH 2 CH 2 CH 2 , preferably CH 2 CH 2 CH 2 .
  • each R 4 and R 5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
  • Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
  • Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
  • Y is substituted with one R 10 .
  • R 10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon.
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety.
  • the cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • Y is oxadiazole or triazole. In some embodiments, Y is ""Q , wherein Ql is O or NR 2 , preferably O
  • R 10 is aryl, arylalkyl, heteroaryl, or hctcroarylalkyl, for example optionally substituted with one or more R 16 .
  • R 10 is substituted with one R 16 , such as halo (e.g., fluoro or chloro) or alkoxy.
  • R 1 is hydrogen or alkyl, for example unsubstituted or substituted with one R 6 ; n is 0 or 1 ; k' is a bond; and
  • R 2 and R 3 each independently hydrogen or Ci-C 6 alkyl
  • R 4 and R 5 are each independently hydrogen or alkyl
  • Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R 10 ; each R 10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroaryl, heteroarylalkyl, -OR 11 , -
  • R 1 ] and R 1 r are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
  • R 12 and R 12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R 12 and R 12 taken together can be cyclized to form -(CH 2 ) q X(CH 2 )s-; wherein each R 12 and R 12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR 11 , alkoxy, heterocycloalkyl, -NR 11 C(O)NR 11 R 11 ', -C(O)NR 11 R 11' , -NR 11 C(O
  • X is CR 11 R 11' , O, S, S(O), S(O) 2 , or NR 11 ;
  • m is an integer between 1 and 6;
  • p is an integer from O to 5;
  • q and s are each independently an integer between 1 and 3; and
  • w is an integer between 0 and 5.
  • n is 0 or 1;
  • k' is a bond; and
  • R 1 is alkyl, for example unsubstituted or substituted with one R 6 .
  • n is 0 and k' is a bond.
  • Exemplary R 1 moieties include methyl, and ethyl.
  • Preferred R 1 moieties include methyl,
  • R 1 is unsubstituted methyl or methyl or ethyl substituted with C(O)N (R 3 )2.
  • n is 0 or 1; k' is a bond; and R 1 is alkyl, for example unsubstituted or substituted with one R 6 .
  • R 1 can be a branched alkyl such as one of the following .
  • n is 0 and k' is a bond, and R 1 and R 3 are both methyl.
  • R 1 is hydrogen
  • A is CH 2 CH 2 or CH 2 CH 2 CH 2 , preferably CH 2 CH 2 CH 2 .
  • each R 4 and R 5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
  • Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
  • Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
  • Y is substituted with one R 10 .
  • R 10 can be positioned, for example, 1 ,3 relative to the point of attachment of Y to the adjacent chain carbon or 1 ,2 relative to the point of attachment of Y to the adjacent chain carbon.
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety.
  • the cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • Y is oxadiazole or triazole.
  • Y is > wherein Ql is O or NR 2 , preferably O or
  • R 10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R 16 .
  • R 10 is substituted with one R 16 , such as halo (e.g., fluoro or chloro) or alkoxy.
  • the compounds has a formula (Ib)
  • R 1 is hydrogen or alkyl
  • A is CH 2 , CH 2 CH 2 , or CH 2 CH 2 CH 2
  • R 2 is hydrogen or C 1 -C 3 alkyl
  • R 4 and R 5 are each independently hydrogen or alkyl
  • Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R 10 ; each R 10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR 11 , - NR 11 R 11' , -CF 3 , -SOR 12 , -SO 2 R 12 , -OC(O)R 11 , -SO 2 NR 12 R 12' , -(CH 2 ) m R 14 or R 15 ; each of which is optionally independently substituted with 1-3 R 16 ;
  • R 11 and R 11 are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
  • R 12 and R are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R 12 and R 12 taken together can be cyclized to form -(CH 2 ) q X(CH 2 ) s -; wherein each R 12 and R 12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR 11 , alkoxy, heterocycloalkyl, -NR 11 C(O)NR 11 R 11' , -C(O)NR 11 R 11' , -NR 11 C(O)R 11' , -CN, oxo, -NR 11 SO 2 R 11' , -OC(O)R 11 , -SO 2 NR 11 R 11' ,
  • each R 15 is independently -(CH 2 ) P N(R 12 )C(O)R 12' , -(CH 2 ) P CN, - (CH 2 )pN(R 12 )C(O)OR 12' , -(CH 2 )pN(R 12 )C(O)NR 12 R 12' , -(CH 2 ) P N(R 12 )SO 2 R 12 , - (CH 2 ) P SO 2 NR 12 R 12' , -(CH 2 ) P C(O)NR 12 R 12' , -(CH 2 ) P C(O)OR 12 , -(CH 2 ) P OC(O)OR 12 , - (CH 2 )pOC(O)R 12 , -(CH 2 )pOC(O)R 12 , -(CH 2 ) P OC(O)NR 12 R 12' , -(CH 2 ) P N(
  • X is CR 11 R 11 ' , O, S, S(O), S(O) 2 , or NR 11 ;
  • m is an integer between 1 and 6;
  • p is an integer from O to 5;
  • q and s are each independently an integer between 1 and 3; and
  • w is an integer between 0 and 5.
  • A is CH 2 CH 2 or CH 2 CH 2 CH 2 , preferably CH 2 CH 2 CH 2 .
  • each R 4 and R 5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
  • Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
  • Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
  • Y is substituted with one R 10 .
  • R 10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon.
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl. In some embodiments, R 10 is substituted with 1-3 R 16 . In some embodiments, R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety.
  • the cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • Y is oxadiazole or triazole.
  • Y is NH.
  • R 10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R 16 .
  • R 10 is substituted with one R 16 , such as halo (e.g., fluoro or chloro) or alkoxy.
  • R 1 and R 3 together form a heterocyclic ring such as a pyrrolidine or an azetidine ring (The heterocyclic ring can be unsubstituted or substituted, for example, with 1-2 R 6 .); n is 0 or 1 ; k' is a bond;
  • R 2 hydrogen or C 1 -Ce alkyl, preferably hydrogen
  • x and y are each independently 0-6;
  • R 4 and R ⁇ arc each independently hydrogen or alkyl;
  • Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R 10 ;
  • each R 10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroarylalkyl, heteroarylalkyl, -OR 11 , -
  • R 1 ' and R 1 ' are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
  • R 12 and R 12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R 12 and R 12 taken together can be cyclized to form -(CH 2 ) q X(CH 2 ) s -; wherein each R 12 and R 12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR 11 , alkoxy, heterocycloalkyl, -NR 11 C(O)NR 11 R 11 ', -C(O)NR 11 R 11' , -NR 11 C
  • X is CR 11 R 11' , O, S, S(O), S(O) 2 , or NR 11 ;
  • m is an integer between 1 and 6;
  • p is an integer from O to 5;
  • q and s are each independently an integer between 1 and 3; and
  • w is an integer between 0 and 5.
  • A is CH 2 CH 2 or CH 2 CH 2 CH 2 , preferably CH 2 CH 2 CH 2 .
  • each R 4 and R 5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
  • Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
  • Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl. In some embodiments, R 10 is substituted with 1-3 R 16 . In some embodiments, R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety.
  • the cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • Y is oxadiazole or triazole.
  • Y is "Q , wherein Ql is O or NR 2 , preferably O NH.
  • R 10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R 16 .
  • R 10 is substituted with one R 16 , such as halo (e.g., fluoro or chloro) or alkoxy.
  • the compounds has a formula (Ic) ⁇ ] R A
  • n is O, 1, 2, 3, or 4; preferably 1 or 2;
  • A is CH 2 , CH 2 CH 2 , or CH 2 CH 2 CH 2 ;
  • R 4 and R 5 are each independently hydrogen or alkyl;
  • Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with l-4 R 10 ; each R 10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, heteroarylalkyl, -OR 11 , -
  • R 11 and R 11 are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
  • R 12 and R 12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R 12 and R 12' taken together can be cyclized to form -(CH 2 ) q X(CH 2 ) s -; wherein each R 12 and R 12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR 11 , alkoxy, heterocycloalkyl, -NR 11 C(O)NR 11 R 11' , -C(O)NR 11 R 11' , -NR 11 C(O)
  • each R 16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH 2 ) P NR 11 C(O)NR 11 R 11' , -(CH 2 ) P C(O)NR 11 R 11' , -(CH 2 ) P NR 11 C(O)R 11' , -CN, - (CH 2 ) P NR 11 SO 2 R 11' , -(CH 2 ) P OC(O)R 1 ⁇ -(CH 2 ) P SO 2 NR 11 R 11' , -(CH 2 ) P SOR 12 , -(CH 2 ) P SO 2 R 12 , -(CH 2 ) P NR 11 R 11 or - (CH 2 ) P OCH 2 C(O)N(R 12 )(CH 2 ) m OH; each R 16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH 2 ) P
  • X is CR 11 R 11' , O, S, S(O), S(O) 2 , or NR 11 ;
  • m is an integer between 1 and 6;
  • p is an integer from O to 5;
  • q and s are each independently an integer between 1 and 3; and
  • w is an integer between O and 5.
  • A is CH 2 CH 2 or CH 2 CH 2 CH 2 , preferably CH 2 CH 2 CH 2 .
  • each R 4 and R 5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
  • Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
  • Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
  • Y is substituted with one R 10 .
  • R 10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon.
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroary alkyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • R 10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety.
  • the cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
  • Y is oxadiazole or triazole.
  • Y is "Q , wherein Ql is O or NR 2 , preferably O or
  • R 10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R 16 .
  • R 10 is substituted with one R 16 , such as halo (e.g., fluoro or chloro) or alkoxy.
  • the invention features a compound of formula (IV)
  • R 2 is hydrogen, C I -C O alkyl, C 2 -C ⁇ alkenyl, or C 2 -C O alkynyl;
  • A' is heterocyclyl; optionally substituted with 1-3 R 9 ;
  • Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R 10 ; each R 6 is independently halo, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl, heteroaryl, alkoxy, haloalkyl, haloalkyloxy, haloalkylthio, acetyl, cyano, nitro, hydroxy, oxo, C(O)OR 2 , OC(O)R 2 , N(R 3 ) 2 , C(O)N(R 3 ) 2 , NR 3 C(O)R 2 , or SR 2 ;
  • R 9 is halo, alkyl, cyclyl, heterocyclyl, aryl, heteroaryl, alkoxy, haloalkyl, haloalkyloxy, haloalkylthio, acetyl, cyano, nitro, hydroxy, oxo, C(O)OR 2 , OC(O)R 2 , N(R 2 ) 2 , C(O)N(R 2 ) 2 , NR 2 C(O)R 2 , SR 2 ; each R 10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl,
  • R 11 and R 11 are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
  • R 12 and R 12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R 12 and R 12 taken together can be cyclized to form -(CH 2 ) q X(CH2)sS wherein each R 12 and R 12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR 11 , alkoxy, heterocycloalkyl, -NR 11 C(O)NR 11 R 11' , -C(O)NR 11 R 11' , -NR 11 C(O)R 11' , -CN, oxo, -NR 11 SO 2 R 11' , -OC(O)R 11 , -SO 2 NR 11 R 11' , -SOR
  • each R 15 is independently heterocycloalkyl, heteroaryl, -CN, - (CH 2 ) p N(R 12 )C(O)R 12' , -(CH 2 ) p CN, -(CH 2 ) P N(R 12 )C(O)OR 12' , - (CH 2 )pN(R 12 )C(O)NR 12 R 12' , -(CH 2 ) P N(R 12 )SO 2 R 12 , -(CH 2 ) P SO 2 NR 12 R 12' , - (CH 2 ) P C(O)NR 12 R 12' , -(CH 2 ) P C(O)OR 12 , -(CH 2 ) P OC(O)OR 12 , -(CH 2 ) P OC(O)OR 12 , -(CH 2 ) P OC(O)R 12 , -(CH 2 ) P OC(O)R 12 , -(CH 2 ) P
  • each R 16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH 2 ) P NR 11 C(O)NR 1 1 R 1 r , -(CH 2 ) p C(0)NR n R n' , -(CH 2 ) P NR 11 C(O)R 11' , -CN, - (CH 2 ) P NR 11 SO 2 R 11' , -(CH 2 ) P OC(O)R 1 ⁇ -(CH 2 ) P SO 2 NR 11 R 11' , -(CH 2 ) P SOR 13 , - (CH 2 ) p C00H or -(CH 2 ) P
  • X is CR 11 R 11' , O, S, S(O), S(O) 2 , or NR 11 ;
  • m is an integer between 1 and 6;
  • p is an integer from O to 5.
  • q and s are each independently an integer between 1 and 3; and
  • w is an integer between 0 and 5.
  • the compound of formula (IV) comprises an enriched preparation of formula (IV)
  • the compound of formula (IV) comprises an enriched preparation of formula (IV")
  • A' is a 5 or 6 membered heterocyclyl.
  • the 5 or 6 membered heterocyclyl includes at least two nitrogen atoms.
  • A' is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • A' is substituted with one R 9 , for example, N(R 2 )2.
  • R 1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl.
  • n is 1; k' is O; and
  • R 1 is arylalkyl.
  • R 1 can be phenylmethyl.
  • n is 2; k' is a bond; and
  • R 1 is aryl
  • Y is a monocyclic heteroaromatic moiety, for example, a nitrogen containing heteraromatic moiety, such as a nitrogen containing 5 membered heteraromatic moiety.
  • Y is a heterocyclic moiety containing at least two heteroatoms, for example, a 5 membered heterocyclic moiety containing at least two heteroatoms or a heterocyclic moiety containing at least 3 heteroatoms.
  • Y is substituted with 1 R 10 .
  • the R 10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or can be positioned, for example, 1,2 relative to the point of attachment of Y to the adjacent chain carbon.
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, or thiophenyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, imidazolyl, benzoxazolyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, or methoxy.
  • Y is oxadiazole or triazole.
  • the invention features a compound of formula (V),
  • each Q 1 , Q 2 , Q 3 and Q 4 together with the carbon to which they are attached form a heteroaryl moiety, and each Q 1 , Q 2 , Q 3 and Q 4 is independently S, O, N, CR 2 , CR 10 , NR 2 , or NR 10 .
  • the compound of formula (V) comprises an enriched preparation of formula (V)
  • the compound of formula (V) comprises an enriched preparation of formula (V") formula (V").
  • Q 1 and Q 4 are each independently S, O, N, or NR 10 . In some embodiments, Q 1 and Q 3 are each independently S, O, N, or NR 10 .
  • Q 2 is CR 2 or CR 10 .
  • Q 2 is S, O, N, or NR 10 .
  • At least one of Q 2 or Q 3 is CR 2 or CR 10 .
  • At least two of Q 1 , Q 2 , Q 3 , or Q 4 is S, O, N, or NR 10 .
  • Q 1 , Q 2 , and Q 3 are each independently S, O, N, or NR 10 .
  • Q 1 is NR 10 .
  • one of Q 2 , Q 3 , or Q 4 is CR 2 '
  • Q 2 is CR 10 .
  • Q 3 is CR 2' In some embodiments, Q 1 , Q 2 , Q 3 and Q 4 together form
  • Q 1 is NR 2 .
  • Q 1 , Q 2 , Q 3 and Q 4 together form
  • Q 1 is NR 10 .
  • the invention features a compound of formula (Vl),
  • Z 1 , Z 2 , Z 3 , Z 4 , and Z 5 together form an aryl or heteroaryl moiety, and each Z 1 , Z 2 , Z 3 , and Z 5 is independently N, CR 10 , or CR 2 .
  • the compound of formula (VI) comprises an enriched preparation of a compound of formula (VF).
  • the compound of formula (VI) comprises an enriched preparation of a compound of formula (VI").
  • one of Z 1 , Z 2 , Z 3 , Z 4 , and Z 5 is N. In some embodiments, two of Z 1 , Z 2 , Z 3 , Z 4 , and Z 5 are N.
  • three of Z 1 , Z 2 , Z 3 , Z 4 , and Z 5 is N.
  • two of Z 1 and Z 2 are N.
  • two of Z 1 and Z 3 are N.
  • two of Z 1 and Z 4 are N.
  • two of Z 1 , Z 3 , and Z 5 are N.
  • the compound is a compound of formula (IV), wherein Y is substituted with a single substituent R 10 .
  • R 10 can be aryl or heteroaryl, optionally substituted with up to three independent R 16 .
  • R 10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, or thiophenyl. In some embodiments, R 10 is substituted with 1-3 R 16 . In some embodiments, R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, or methoxy.
  • R 10 is a bicyclic heteroaryl, for example indolyl, imidazolyl, benzoxazolyl, or benzthiazolyl.
  • R 10 is substituted with 1-3 R 16 .
  • R 16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, or methoxy.
  • R 10 is R 15 .
  • Y is substituted with a second R 10 , for example an alkyl, halo or alkoxy.
  • the invention features a pharmaceutically acceptable salt comprising a compound of any of the formulae described herein.
  • 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 or delaying the onset of diabetes comprising administering to a subject a compound of any of the formulae described herein.
  • 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.
  • 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
  • a compound described herein can be administred 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 invention features a compound listed in Table 1.
  • 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), (II), (III), (IV), (V), or (VI).
  • 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), (II), (III), (IV), (V), or (VI).
  • 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%.
  • enriched preparation is enriched for a selected stereoconfiguration of one, two, three or more selected stereocenters within the subject compound.
  • Exemplary selected stereocenters and exemplary stereoconf ⁇ gurations thereof can be selected from those provided, herein, e.g., in an example described herein, e.g., those described in the Table below.
  • 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.
  • a preparation of a compound disclosed herein is enriched for isomers (subject isomers) which are diastereromers of the 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), (II), (III), (IV), (V), or (VI).
  • 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%.
  • Diastereromer 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.
  • 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 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.
  • 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 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.
  • the subject is a mammal, e.g., a human, a primate, a dog, a cat, a racing, purebred, or an agricultural mammal.
  • the subject is overweight or obese.
  • 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.
  • 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,
  • 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.
  • 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.
  • 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 compound is administered orally, or parenterally, e.g., by injection, and so forth.
  • the compound is administered at a plurality of intervals, e.g., regular intervals.
  • 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.
  • GHS-R e.g., resistin, leptin, or adiponectin
  • 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 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.
  • 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 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.
  • 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.
  • 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.
  • 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.
  • 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).
  • 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.
  • the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
  • the additional therapeutic agent is an insulin preparation, an oral hypoglycemic, a peroxisome proliferator-activated receptor gamma agonist, a nitrate, a nitrite, a ⁇ -blocker, a calcium channel antagonist, a peripheral vasodilator, a cinchona alkaloid, an amide, a quaternary ammonium salt, a diidobenzyloxyethylamine, a hydantoin, a diuretic, a vasodilator, a peripheral sympatholytic, a central ⁇ 2 -sympathomimetic, an angiotensin-converting enzyme inhibitor, an angiotensin II-receptor antagonist, a HMG CoA reductase inhibitor, a fibrate
  • the invention features a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI, and an additional therapeutic agent.
  • the additional therapeutic agent is an insulin preparation, an oral hypoglycemic, a peroxisome proliferator-activated receptor gamma agonist, a nitrate, a nitrite, a ⁇ -blocker, a calcium channel antagonist, a peripheral vasodilator, a cinchona alkaloid, an amide, a quaternary ammonium salt, a diidobenzyloxyethylamine, a hydantoin, a diuretic, a vasodilator, a peripheral sympatholytic, a central ⁇ 2 -sympathomimetic, an angiotensin-converting enzyme inhibitor, an angiotensin II-receptor antagonist, a HMG CoA reductase inhibitor,
  • the invention features a method of treating a disorder in a subject.
  • the method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
  • the ghrelin antagonist is co-administered with the additional therapeutic agent.
  • the ghrelin antagonist and the additional therapeutic agent are administered in a single dosage form.
  • the ghrelin antagonist and the additional therapeutic agent are provided in amounts so as to provide a synergistic effect.
  • the ghrelin antagonist and the additional therapeutic agent are administered one time daily.
  • the ghrelin antagonist and the additional therapeutic agent are administered twice daily.
  • the ghrelin antagonist and the additional therapeutic agent are administered three times daily. In an embodiment, the ghrelin antagonist and the additional therapeutic agent are administered within 1 hour before or two hours after a meal. In an embodiment, the ghrelin antagonist and the additional therapeutic agent are administered without regard to food.
  • the disorder is obesity. In an alternative embodiment, the disorder is a combination of disorders. In a further alternative embodiment, the combination of disorders is obesity and an additional disorder.
  • the additional therapeutic agent is an insulin preparation, an oral hypoglycemic, a peroxisome proliferator-activated receptor gamma agonist, a nitrate, a nitrite, a ⁇ - blocker, a calcium channel antagonist, a peripheral vasodilator, a cinchona alkaloid, an amide, a quaternary ammonium salt, a diidobenzyloxyethylamine, a hydantoin, a diuretic, a vasodilator, a peripheral sympatholytic, a central ⁇ 2 -sympathomimctic, an angiotcnsin- converting enzyme inhibitor, an angiotensin ⁇ -rece ⁇ tor antagonist, a HMG CoA reductase inhibitor, a f ⁇ brate, an antiarrhythmic, a cardiac glycoside, a heparin, a coumarin derivative, a histone
  • the insulin preparation is a short acting insulin, regular insulin, semilente insulin, intermediate acting insulin, lente insulin, or long acting insulin.
  • the oral hypoglycemic is a sulfonylurea, a mcglitinidc, ⁇ -glucosidasc inhibitor, or an insulin sensitizer.
  • the sulfonylurea is tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, or glimepiride.
  • the meglitinide is mitiglinide, repaglinide or nateglinide.
  • the ⁇ - glucosidase inhibitor is acarbose, miglitol, voglibose (N-l(l,3-dihydroxy-2-propyl) valiolamine, and N-substituted derivatives thereof, and N-substituted pseudo-amino sugars.
  • the insulin sensitizer is a biguanide, or a thiazolidinedione.
  • the biguanide is metformin.
  • the thiazolidinedione is pioglitazone or rosiglitazone.
  • the peroxisome proliferator-activated receptor gamma agonist is a thiazolidinedione.
  • the nitrate is nitroglycerin, isorbide, isorbide-5-mononitrate, or erythrityl tetranitrate.
  • the nitrite is amyl nitrate.
  • the ⁇ -blocker is propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, carteolol, oxprenolol, penbutolol, levatol, medroxalol, bucindolol, levobunolol, metipranolol, bisoprolol, nebivolol, betaxolol, celiprolol, sotalol, or propafenone.
  • the calcium channel antagonist is amlodipine, arandipine, bamidipine, bencyclane, benidipine, bepridil, cilnidipine, cinnarizine, clentiazem, diltiazem, efonidipine, elgodipine, etafenone, flunarizine, filodepine, galopamil, isradipine, lacidipine, lercanidipine, lidoflazine, lomerizine, manidipine, mibefradil, nicardipine, nifedipine, nilvadepine, nimodipine, nisoldipine, nitrendipine, penylamine, perhexiline, phendilin, semotiadil, terodiline, or verapamil.
  • the peripheral vasodilator is dipyridamole, nylidrin hydrochloride, or isoxsuprine hydrochloride.
  • the cinchona alkaloids is quinidine or quinine.
  • the amide is procainamide, flecainide, lidocaine, bupivicaine, ropivicaine, or dispyramide.
  • the diidobenzyloxyethylamine is amiodarone.
  • the hydantoin is phenytoin.
  • the diuretic is a thiazide diuretic, an osmotic diuretic, a carbonic anhydrase inhibitor, a benzothiadiazide diuretic, a loop diuretic, or a potassium sparing diuretic.
  • the thiazide diuretic is bendrofiuazide, bendroflumethiazide, benzthiazide, benzylhydrochlorothiazide, chlorothiazide, chlorthalidone, cyclopenthiazide, diucardin, diuril, enduron, esidrix, exna, HCTZ, hydrochlorothiazide, hydrodiuril, hydroflumethiazide, bendroflumethiazide, hydromox, hygroton, indapamide, lozol, methylchlorthiazide, metolazone, mykrox, naqua, naturetin, oretic, penflutizide, polythiazide, quinethazone, renese, trichlormethiazide, xipamide, or zaroxolyn.
  • the osmotic diuretic is mannitol, urea, glycerin, or isorbide.
  • the carbonic anhydrase inhibitor is acetazolamide.
  • the benzothiadiazide diuretic is or chlorothiazide.
  • the loop diuretic is azosemide, furosemide, bumetanide, ethacrynic acid, piretanide, or torasemide.
  • the potassium sparing diuretic is triamterene, amiloride, spironolactone, or eplerenone.
  • the vasodilator is bencyclane, cinnarizine, citicoline, cyclandate, cyclonicate, diazoxide, ebumamonine, flunarizine, hydralazine, ibudilast, ifenprodil, lomerizine, minoxidil, naphlole, nikamate, nimodipine, nitroprusside, nofedoline, nosergoline, papaverine, pentifylline, pentoxifylline, phenoxezyl, prostacyclin derivatives, vichizyl, vincamin, or vinpocetine.
  • the peripheral sympatholytic is acebutol, atenolol, betaxolol, bisoprolol, carteolol, carvediolol, doxazosin, guanadrel, guanethidine, labetalol, metoprolol, nadolol, penbutolol, pindolol, prazosin, propanolol, reserpine, terazosin, or timolol.
  • the central ⁇ 2-sympathomimetic is clonidine, guanabenz, guanfacine, or methyldopa.
  • the angiotensin-converting enzyme inhibitor is alacepril, benazepril, captopril, ceranopril, cilazopril, delapril, enalapril, fosinopril, imidapril, lisinopril, mobertpril, moexipril, pentopril, perindopril, quinapril, ramipril, randolapril, spirapril, temocapril, trandolapril, or zofenopril.
  • the angiotensin II- receptor antagonist is candesartan, eprosartan, ibersartan, losartan, telmisartan, or valsartan.
  • the HMG CoA reductase inhibitor is livostatin, simvastatin, pravastatin, fluvastatin, atorvastatin, or cerivastatin.
  • the fibrate is nicotinic acid, bezafibrate, clinofibrate, clofibrate, colestimide, fenof ⁇ brate, gemfibrizil, probucol, or simfibrate.
  • the antiarrhythmic is tocainide, encainide, phenytoin, flecainide, tocainide, propafenone, bretylium, sotalol, ibutilide, dofetilide, bepridil, moricizine, propanolol, esmolol, artilide, clof ⁇ lium, azimilide, dronedarone, ersentilide, ibutilide, tedisamil, valvetilide, verapamil, diltaizem, digitalis, adenosine, nickel chloride, procainaltide, lignocaine, or magnesium ions.
  • the cardiac glycoside is allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, a xanthine formulation, a catecholamine formulation, denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, or colforsin daropate.
  • the xanthine formulation is aminophylline, choline theophylline, diprophylline, or proxphylline.
  • the catecholamine formulation is dopamine, dobutamine, or docarpamine.
  • the heparin agent is heparin sodium, heparin potassium, a low-molecular weight heparin fragment, or a low-molecular weight heparinoid.
  • the low-molecular weight heparin fragment is enoxaparin, dalteparin, tinzaparin, or ardeparin.
  • the low-molecular weight heparinoid is danaparoid.
  • the coumarin derivative is warfarin or dicumarol.
  • the histone deacetylase inhibitor is a class I and/or a class II histone deacetylase inhibitor.
  • the inhibitor of the class I and'or class II histone deacetylase is a small molecular weight carboxylate. In an embodiment, the inhibitor of the class I and/or class II histone deacetylase is a hydroxamic acid. In an embodiment, the inhibitor of the class I and/or class II histone deacetylase is a benzamide. In an embodiment, the inhibitor of the class 1 and/or class 11 histone deacetylase is an epoxyketone. In an embodiment, the inhibitor of the class I and/or class II histone deacetylase is a cyclic peptide. In an embodiment, the inhibitor of the class I and/or class II histone deacetylase is a hybrid molecule.
  • the inhibitor of the class I and'Or class II histone deacetylase is Suberoylanilide Hydroxamic Acid (SAHA (e.g., MK0683, vorinostat) and other hydroxamic acids), Suberoyl bis-hydroxamic Acid, BML-210, Depudecin (e.g., (-)- Depudecin), HC Toxin, Nullscript (4-(l,3-Dioxo-lH,3H-benzo[de]isoqumolm-2-yl)-N- hydroxybutanamide), Phenylbutyrate (e.g., sodium phenylburyrate) and Valproic Acid (and other short chain fatty acids), Scriptaid, Splitomicin, Suramin Sodium, Trichostatin A (TSA), APHA Compound 8, Apicidin, Sodium Butyrate, pivaloyloxymethyl butyrate (Pivanex, AN- 9), Trapoxin B, Chlamy
  • the SIRT inhibitor is a 2- anilinobenzamide or 2-chloro-5,6,7,8,9,10-hexahydrocyclohepta[b]indole-6, 2-chloro- 5,6,7,8,9,10-hexahydrocyclohepta[b]indole-6-carboxylate, or 2-chloro-5, 6,7,8, 9,10- hexahydrocyclohepta[b]indole-6-carboxamide, or 6-Chloro-2,3 ,4,9-tetrahydro- 1 H- carbazole-1-carboxylic acid amide.
  • the 2-anilinobenzamide is nicotinamide, carba-NAD + , EX-527, sirtinol, para-sirtinol, JFD00244, splitomicin, HR73, or cambinol.
  • the xanthine oxidase inhibitor is allopurinol.
  • the alkylating agent is altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, or thiotepa.
  • the antitumor antibiotic is bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitoxantrone, or valrubicin.
  • the antimetabolite is capecitabine, cladribine, cytarabine, fludarabine, floxuridine, 5-fluorouracil, gemicabine, hydroxyurea, 6-mercaptopurine, methotrexate, pentostatin, or 6-thioguanine.
  • the mitotic inhibitor is docetaxel, paclitaxel, vinblastine, vincristine, or vinorelbine.
  • the topoisomerase inhibitor is etoposide, irinotecan, teniposide, or topotecan.
  • the enzyme is asparaginase or pagasparaginase.
  • the protein tyrosine kinase inhibitor is imatinib mesylate.
  • the hormonal agent is an adrenocorticoid, an estrogen, a progestin, an antiestrogen, an androgen, or an antiandrogen.
  • the adrenocorticoid is dexamethasone, methylprednisolone, or prednisone.
  • the estrogen is diethylstilbestrol or estradiol.
  • the progestin is medroxyprogesterone or megesterol acetate.
  • the antiestrogen is fulvestrant, tamoxifen, or toremifene.
  • the androgen is testosterone, methyltestosterone, or fluoxymesterone.
  • the antiandrogen is bicalutamide, flutamide, or nilutamide.
  • the aromatase inhibitor is aminoglutethimide, anastrozole, exemestane, or letrozole.
  • the LHRH analogue is leuprolide or goserelin.
  • the biological agent is interferon- ⁇ 2a and 2b, interleukin-2, interleukin-11, the growth factors filgrastim, pegf ⁇ lgrastim, sargramostim, epoetin alfa, darbepoetin alfa, or a monoclonal antibody.
  • the benzodiazepine is diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, and xanax.
  • the opioid-receptor antagonist is tramadol or naltrexone.
  • the antidepressant is a selective serotonin reuptake inhibitor, a serotonin and norepinephrine reuptake inhibitor, a tetracyclic antidepressant, a norepinephrine and dopamine reuptake inhibitor, a combined reuptake inhibitor and receptor blocker, a tricyclic antidepressant, or a monoamine oxidase inhibitor.
  • the selective serotonin reuptake inhibitor is citalopram, escitalopram, fluoxetine, paroxetine, or sertraline.
  • the serotonin and norepinephrine reuptake inhibitor is duloxetine or venlafaxine.
  • the tetracyclic antidepressant is mirtazapine.
  • the combined reuptake inhibitor and receptor blocker is trazodone, nefazodone, or maprotiline.
  • the tricyclic antidepressant is amitriptyline, amoxapine, desipramine, doxepin, imipramine, nortriptyline, protriptyline, or trimipramine.
  • the monoamine oxidase inhibitor is phenelzine, ranylcypromine, isocarboxazid, or selegiline.
  • the analgesic is paracetamol, a non-steroidal anti-inflammatory drug, a cyclooxygenase-2 inhibitor, an opiate, a corticosteroid, or a topical analgesic.
  • the paracetamol is acetaminophen or indomethacin.
  • the non-steroidal anti-inflammatory drugs comprise, aspirin, ibuprofen, salsalate, magnesium salicylate, or a COX-2 selective inhibitor.
  • the COX -2 selective inhibitor is celecoxib, refecoxib, or valdecoxib.
  • the opiate is codeine, oxycodone, hydrocodone, diamorphine, pethidine, tramadol, or buprenorphine.
  • the corticosteroid is prednisone, prednisolone, cortisone, or hydrocortisone.
  • the topical analgesic is capsaicin.
  • the amphetamine is dextroamphetamine, benzedrine, ritalin, phenmetrazine, phendimetrazine, methamphetamine, diethylpropion, phentermine, sibrutamine, or orlistat.
  • the invention features a method of treating a disorder in a subject.
  • the method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
  • the disorder is obesity.
  • the disorder is a combination of disorders.
  • the combination of disorders is obesity and syndrome X.
  • syndrome X is diabetes.
  • the additional therapeutic agent is an anti-diabetic agent.
  • the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-diabetic agent.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered in a single dosage form.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are provided in amounts so as to provide a synergistic effect.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered one time daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered twice daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered three times daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered within 1 hour before or two hours after a meal. In an embodiment, the ghrelin antagonist, for example a compound of formula 1, II, III, IV, V, or VI and the anti-diabetic agent are administered without regard to food.
  • the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- diabetic agent.
  • a ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti- diabetic agent.
  • the invention features a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-diabetic agent.
  • the solid dosage form includes a tablet or capsule.
  • the solid dosage form includes an enteric coating.
  • the anti-diabetic agent is insulin, tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, glimepiride, repaglinide, nateglinide, acarbose, miglitol, metformin, pioglitazone, or rosiglitazone.
  • the insulin is short acting insulin, regular insulin, semilente insulin, intermediate acting insulin, lente insulin, or long acting insulin.
  • the invention features a method of treating a disorder in a subject. The method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
  • the disorder is obesity.
  • the disorder is a combination of disorders.
  • the combination of disorders is obesity and cardiovascular disease.
  • the additional therapeutic agent is a cardiovascular disease treating agent.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI is co-administered with a cardiovascular disease treating agent.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered in a single dosage form.
  • the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are provided in amounts so as to provide a synergistic effect.
  • the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered one time daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered twice daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered three times daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered within 1 hour before or two hours after a meal. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered without regard to food.
  • the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent.
  • the pharmaceutical composition is in a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent.
  • the solid dosage form includes a tablet or capsule.
  • the tablet or capsule includes an enteric coating.
  • the cardiovascular treating agent is, allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, aminophylline, choline theophylline, diprophylline, proxphylline, dopamine, dobutamine, docarpamine, denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, colforsin daropate, amyl nitrite, nitroglycerin, propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, carteolol, oxprenol
  • the invention features a method of treating a disorder in a subject.
  • the method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
  • the disorder is obesity.
  • the disorder is a combination of disorders.
  • the combination of disorders is obesity and cancer.
  • the additional therapeutic agent is an anti-cancer agent.
  • the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-cancer agent.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anticancer agent are administered in a single dosage form.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are provided in amounts so as to provide a synergistic effect.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anticancer agent are administered one time daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered twice daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer treating agent are administered three times daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered within 1 hour before or two hours after a meal.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered without regard to food.
  • the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anticancer agent.
  • the pharmaceutical composition is in a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent.
  • the solid dosage form includes a tablet or capsule.
  • the tablet or capsule includes an enteric coating.
  • the anticancer agent is, altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, thiotepa, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitoxantrone, valrubicin, capecitabine, cladribine, cytarabine, fludarabine, floxuridine, 5-fluorouracil, gemicabine, hydroxyurea, 6-mercaptopurine, methotrexate, pentostatin, 6-thioguanine, docetaxel, paclitaxel, vin
  • the invention features a method of treating a disorder in a subject.
  • the method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
  • the disorder is obesity.
  • the disorder is a combination of disorders.
  • the combination of disorders is obesity and addiction.
  • the additional therapeutic agent is an anti-addictive agent.
  • the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-addictive agent.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered in a single dosage form.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti- addictive agent are provided in amounts so as to provide a synergistic effect.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered one time daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti- addictive agent are administered twice daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive treating agent are administered three times daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered within 1 hour before or two hours after a meal.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti- addictive agent are administered without regard to food.
  • the invention features a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent.
  • the pharmaceutical composition is in a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- addictive agent.
  • the solid dosage form includes a tablet or capsule.
  • the tablet or capsule includes an enteric coating.
  • the anti-addictive agent is, nicotine replacement, diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, xanax, propanolol, clonidine, bupropion hydrochloride, naltrexone, citalopram, fluoxetine, zimelidine, sertraline, paroxetine, disulfiram, naltrexone, nalmefene, acamprosate, buspirone, or ondansetron.
  • the invention features a method of treating a disorder in a subject.
  • the method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
  • the disorder is obesity.
  • the disorder is a combination of disorders.
  • the combination of disorders is obesity and osteoarthritis.
  • the additional therapeutic agent is an anti- osteoarthritic agent.
  • the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-osteoarthritic agent.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or Vl and an anti-osteoarthritic agent are administered in a single dosage form.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are provided in amounts so as to provide a synergistic effect.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered one time daily.
  • the ghrelin antagonist for example a compound of formula I, II,
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered twice daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered three times daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered within 1 hour before or two hours after a meal.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered without regard to food.
  • the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- osteoarthritic agent.
  • a ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI
  • an anti- osteoarthritic agent for example a compound of formula I, II, III, IV, V, or VI.
  • the pharmaceutical composition is in a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III,
  • the solid dosage form includes a tablet or capsule.
  • the tablet or capsule includes an enteric coating.
  • the antiosteoarthritic is, acetaminophen, salsalate, magnesium salicylate, aspirin, diclofenac, etodolac, ibuprofen, naproxen, nabumeton, sulindac, tolmetin, celecoxib, refecoxib, valdecoxib, tramadol, codeinek, oxycodone, capsaicin, corticosteroids, hyaluronic acid derivatives, glucosamine, chondroitin, or S- adenosyl-methionine.
  • the invention features a method of treating a disorder in a subject.
  • the method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
  • the additional therapeutic agent is a weight loss promoting agent.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI is co-administered with a weight loss promoting agent.
  • the ghrelin antagonist, for example a compound of formula I, II, 111, IV, V, or VI and a weight loss promoting agent are administered in a single dosage form.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are provided in amounts so as to provide a synergistic effect.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered one time daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered twice daily.
  • the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered three times daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or Vl and a weight loss promoting agent are administered within 1 hour before or two hours after a meal.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered without regard to food.
  • the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent.
  • the pharmaceutical composition is in a solid dosage form inlcuding a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent.
  • the solid dosage form includes a tablet or capsule.
  • the tablet or capsule includes an enteric coating.
  • the weight loss promoting therapeutic agent is, dexfenfluramine, amphetamine, methamphetamine, phenmetrazine, phendimetrazine, diethylpropion, phentermine, sibutramine, or orlistat.
  • the invention features a method of treating a disorder in a subject.
  • the method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
  • the disorder is obesity.
  • the disorder is a combination of disorders.
  • the combination of disorders is obesity and gout.
  • the additional therapeutic agent is an anti-gout agent.
  • the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or Vl and an anti-gout agent are administered in a single dosage form.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are provided in amounts so as to provide a synergistic effect.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered one time daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered twice daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered three times daily.
  • the ghrelin antagonist for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered within 1 hour before or two hours after a meal.
  • the ghrelin antagonist for example a compound of formula 1, 11, III, IV, V, or VI and an anti-gout agent are administered without regard to food.
  • the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- gout agent.
  • the pharmaceutical composition is in a solid dosage form inlcuding a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent.
  • the solid dosage form includes a tablet or capsule.
  • the tablet or capsule includes an enteric coating.
  • the anti-gout agent is, vorinostat, zolinza, SAHA, depsipeptide, MS-275, MGCD0103, PXDlOl, baceca, savicol, LBH589, PCI-24781, or ITF2357.
  • halo refers to any radical of fluorine, chlorine, bromine or iodine.
  • 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.
  • lower alkyl refers to a Cl-C 8 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.
  • alkoxy refers to an -O-alkyl radical.
  • 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. For example, 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. For example, 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 aromatic ring system 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.
  • amino acid refers to a molecule containing both an amino group and a carboyxl group.
  • Suitable amino acids include, without limitation, both the D- and L- isomers of the 20 naturally occurring amino acids found in peptides (e.g., A, R, N, C, D, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y, V (as known by the one letter abbreviations)) as well as unnaturally occurring amino acids prepared by organic synthesis or other metabolic routes.
  • amino acid side chain refers to any one of the twenty groups attached to the ⁇ -carbon in naturally occurring amino acids.
  • amino acid side chain for alanine is methyl
  • amino acid side chain for phenylalanine is phenylmethyl
  • amino acid side chain for cysteine is thiomethyl
  • amino acid side chain for aspartate is carboxymethyl
  • amino acid side chain for tyrosine is 4- hydroxyphenylmethyl, etc.
  • substituted 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- 1 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, FOXO-3, or FOXO-4).
  • PI(3) kinase PTEN phosphatase
  • PI(3,4)P2, 14-3-3 protein protein
  • PI(3,4,5)P3 phosphatidyl inositol kinases e.g., AKT serine/threonine kinase (e.g., AKT-I, AKT-2, or AKT-3)
  • a Forkhead transcription factor such as FOXO-I, FOXO
  • 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.
  • 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 FOXO-4.
  • Exemplary IGF-I Receptor signaling pathway effector can include: GADD45, PA26, Selenoprotein P, Whipl, cyclin G2, andNIP3.
  • 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 constitutive activity of the protein e.g., a receptor
  • 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 (ELlSA), radioimmunoassay (RJA) or polyclonal immunoassay.
  • ELSA enzyme-linked immunosorbent assay
  • RJA 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.
  • 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.
  • IGF 1 levels can be measured and compared to age and gender matched normal controls.
  • 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.
  • Figures 1 and 2 demonstrate that compounds 57 and 94 block ghrelin-induced food intake in mice.
  • Figures 3a and 3b demonstrate how administration of compounds 57 and 94 resulted in decreased body weight in mice. No apparent effect on food intake was observed.
  • Figures 4a-4f demonstrate improvement in insulin sensitivity in mice dosed with compounds 57 and 94.
  • Figure 5 demonstrates improved plasma HbAIc levels and improved cholesterol levels at 28 days in mice dosed with compound 57.
  • Figure 6 demonstrates improved plasma HbAIc levels, improved cholesterol levels and improved triglyceride levels at 28 days in mice dosed with compound 94.
  • Figure 7 demonstrates improved fasting blood glucose at 14 days in mice dosed with compound 57.
  • 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.
  • A refers to a compound having antagonist activity with a Ki ⁇ 100 nM in a cell based assay.
  • B refers to a compound having antagonist activity with a Ki between 100 nM and 500 nM in a cell based assay.
  • C refers to a compound having antagonist activity with a Ki between 500 nM and 1000 nM in a cell based assay.
  • D refers to a compound having antagonist activity with Ki, ⁇ 1000 nM in a cell-based assay.
  • GHS-R refers to other exemplary compounds.
  • Representative compounds that modulate GHS-R include the compounds of formulas
  • Y is a 5 membered heteroaromatic moiety substituted with 1 or 2 substituents as described herein.
  • Exemplary Y moieties are reproduced below.
  • any atom, including the hydrogens depicted on the nitrogen atoms, can be substituted with R 10 .
  • the heteroaryl moiety includes 1 or 2 R 10 substituents.
  • R 10 is aryl, arylalkyl, or R 15 . When two R 10 substituents are included, in some embodiments, one R 10 is R 15 and the second R 10 is a different substituent, such as alkyl, alkoxy, halo, etc.
  • R 1 is an aryl moietiy such as a phenyl moiety, for example unsubstituted or substituted aryl moiety.
  • R 1 is a heteroaryl moiety such as an indole moiety.
  • K is an oxygen or a bond.
  • a and R 4 and R 5 can be chosen to vary the compound's type of interaction with GHS-
  • R for example, in some instances where R 4 and R 5 are both hydrogen, the compound is an agonist of GHS-R. In other instances where R 4 and R 5 are both independently alkyl, the compound is an antagonist of GHS-R.
  • compositions 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.
  • additional therapeutic compound e.g., an anti-hypertensive compound or a cholesterol lowering compound
  • stable 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).
  • a Y moiety or other ring corresponding to a Y moiety, can be synthesized onto an amino acid or amino acid type starting material as depicted in schemes A and B and B' below.
  • PG is a nitrogen protecting group.
  • the nitrogen protected amino acid is reacted with a N- hydroxy imidamide (amidoxime) moiety (which is prepared by reacting a cyano containing moiety with hydroxylamine) to produce an oxadiazole containing moiety.
  • the resulting compound can be further manipulated to form a compound of formula (I) by removing the nitrogen protecting group and reacting the resulting moiety with an activated sulfone, such as a sulfonyl chloride as depicted below.
  • Scheme B depicts the formation of a triazole containing moiety which can be further reacted in a manner similar to the oxadiazole containing moiety to form a compound of formula (I).
  • the triazole precursor moiety can be prepared in a variety of manners, for example, by reacting a cyano containing moiety with a hydrazine hydrate (to form the intermediate amidrazone) as depicted in scheme B.
  • the triazole precursor moiety can be prepared as shown in scheme B' by reacting a nitrile moiety (e.g., an arylnitrile or benzylnitrile) with a dialkyl dithiophosphate moiety such as diethyl dithiophosphatc to provide an thioimidatc, which is reacted with a acyl hydrazidc moiety to provide the triazole precursor.
  • the acyl hydrazide moiety can be prepared, for example, by reacting a carboxylic acid or derivative thereof with hydrazine.
  • a compound of formula (I) can be prepared by first reacting an activated sulfone moiety (e.g., a sulfonyl chloride) with an amino acid moiety or protected amino acid, as depicted in Scheme C below.
  • an activated sulfone moiety e.g., a sulfonyl chloride
  • the free carboxyl moiety can then be further manipulated to produce a compound of formula (I).
  • the free carboxyl moiety can be reacted with a compound of formula (X) or (XI) above to form an oxadiazole or triazole containing compound of formula (I) in a manner similar to that described in schemes A, B and B' above.
  • solid support refers a material to which a compound is attached to facilitate identification, isolation, purification, or chemical reaction selectivity of the compound.
  • 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, Czaraik, A.W., Curr. Opin. Chem. Bio., (1997) 1, 60).
  • functional groups such as amino, hydroxy, carboxy, or halo groups
  • solid phase peptide refers to an amino acid, which is chemically bonded to a resin (e.g., a solid support).
  • Resins are generally commercially available (e.g., from SigmaAldrich). Some examples of resins include Rink-resins, Tentagel S RAM, MBHA, and BHA-resins.
  • 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. AH crystal forms of the compounds described herein are expressly included in the present invention.
  • 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.
  • compositions of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • 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 ⁇ 1 salts.
  • alkali metal e.g., sodium
  • alkaline earth metal e.g., magnesium
  • ammonium e.g., sodium
  • N ⁇ alkyl ⁇ 1 salts e.g., sodium
  • alkali metal e.g., sodium
  • alkaline earth metal e.g., magnesium
  • ammonium e.g., sodium
  • N ⁇ alkyl ⁇ 1 salts e.g., sodium
  • alkaline earth metal e.g., magnesium
  • ammonium e.g., sodium
  • N ⁇ alkyl ⁇ 1 salts e.g., sodium
  • alkaline earth metal e.g., magnesium
  • ammonium e.g., sodium
  • N ⁇ alkyl ⁇ 1 salts e.g., sodium
  • 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 2+ 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.
  • 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 10 cells per well so that binding is assayed in the range of about 5 - 8 % binding of the radioactive ligand.
  • 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.
  • 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.
  • Kd and Ki dissociation and inhibition constants
  • IP inositol phosphates
  • a mammalian cell e.g., a Cos-7 cells.
  • Other tissue culture cells, Xenopus oocytes, and primary cells can also be used.
  • COS-7 cells are incubated for 24 hours with 5 ⁇ Ci of [ 3 H]-my ⁇ -inositol 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 MgSO 4 , 1 mM CaCl 2 , 10 mM glucose, 0.05 % (w/v) bovine serum; and incubated in 0.5 ml buffer supplemented with 10 mM LiCl at 37 0 C for 30 min.
  • 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 trans fected with a mixture of pFA2-CREB and pFR-Luc reporter plasmid (PathDetectTM CREB trans-Reporting System, Stratagene) and nucleic acid encoding GHS.
  • PathDetectTM CREB trans-Reporting System Platagene
  • the assay is ended by washing the cells twice with PBS and adding lOO ⁇ l luciferase assay reagent (LucLiteTM, Packard Bioscience). Luminescence is measured (e.g., as relative light units (RLU)) using in a luminometer such as the TopCounterTM (Packard Bioscience) for 5 sec.
  • lOO ⁇ l luciferase assay reagent Luciferase assay reagent
  • Luminescence is measured (e.g., as relative light units (RLU)) using in a luminometer such as the TopCounterTM (Packard Bioscience) for 5 sec.
  • GHS-R regulated genes 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 EC5 0 values can be determined by nonlinear regression, e.g., using the Prism 3.0 software (GraphPad Software, San Diego).
  • Exemplary in vivo assays include the fast-refeeding assay described in Example 1 and as follows.
  • 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
  • the compound of interest can also be evaluated in other experiments.
  • 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.
  • the animal is evaluated, e.g., for feeding behavior, anxiety, or one or more physiological parameters, e.g., a metabolic parameter.
  • each drug can be diluted in 4 ⁇ l of artificial cerebrospinal fluid for injection.
  • mice are anaesthetised 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.
  • 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.
  • 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).
  • mice or other animals provided with the test compound can be analyzed for one or more biological parameters, e.g., metabolic parameters.
  • serum is obtained from blood from the orbital sinus under ether anaesthesia 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.
  • 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 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 cDNA probe (e.g., radioactively, chemically, or fluorescently labeled) for the gene of interest.
  • a labeled cDNA probe e.g., radioactively, chemically, or fluorescently labeled
  • the total integrated densities of hybridization signals can be determined by densitometry.
  • Data can be normalized to a glyceraldehyde 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 K; (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 K D 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.
  • 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.
  • 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 SOD 1-G93 A 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.
  • SOD 1-G93 A transgenic mouse which carries a variable number of copies of the human G93A SOD mutation driven by the endogenous promoter
  • SOD1-G37R transgenic mouse Wang et al., Neuron, 14(6): 1105-16 (1995)
  • SOD1-G85R transgenic mouse Bruijn et al., Neuron, 18(2):327- 38 (1997)
  • C C.
  • 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.
  • IL-I interleukin-1
  • 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 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.
  • MPTP dopaminergic neurotoxin l-methyl-4 phenyl 1,2,3,6- tetrahydropyridine
  • 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.
  • KR-IGF-IR dominant-negative IGF-I receptor
  • 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.
  • 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.
  • the compounds of the formulae described herein can, for example, be administered by injection, intravenously, intraarterial Iy, 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • 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.
  • 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 injectables, 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.
  • 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 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.
  • 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 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.
  • 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 an compound effective to prevent a disorder, or a "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.
  • 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.
  • 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.
  • 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.
  • neurodegenerative 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)
  • DPLA Dentatorubropallidoluysian Atrophy
  • SCAl Spinocerebellar Ataxia 1
  • SCA2 Spinocerebellar Ataxia 2
  • MJD Machado- Joseph Disease
  • SCA6 Spinocerebellar Ataxia 6
  • SCA7 Spinocerebellar Ataxia 7
  • SCA12 Spinocerebellar Ataxia 12
  • 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 timezones, 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.
  • 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, cardiomyopathy, coronary heart disease, venous thromboembolic disease, endocarditis, congenital heart disease, myocardial ischemia, angina, arrhythmia, diseases of the aorta and its branches, diseases of the peripheral vascular system, orthostatic hypotension, hypertension (i.e., elevated blood pressure), hyperlipidemia, arteriosclerosis, artherosclerosis, myocardial infarction, cardiac fibrillation, congestive heart failure, heart failure, and stroke.
  • 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, elevated cholesterol, or elevated blood pressure.
  • 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.
  • a compound described herein for example, a compound of formula I, II, III, IV, V or VI can be used to treat addictive behavior involving, for example, smoking, gambling, and alcoholism.
  • 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-I ⁇ , leptin, and gastrin- releasing peptide. Accordingly, the compounds may increase the discharge rate of the gastric vagal afferent.
  • substance P and derivatives thereof can modulate GHS-R activity.
  • substance P alters feeding activity of mice in the fast refeed assay. Accordingly, substance P and derivatives thereof can be used to modulating an eating or metabolic disorder as well as other disorders described herein.
  • GHS-R is 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 neopla L stic 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,
  • 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.
  • 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 4 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.
  • tumors 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.
  • malignancies e.g., sarcomas, adenocarcinomas, and carcinomas
  • the various organ systems such as those affecting lung, breast, lymphoid, gastrointestinal (e.g., colon), and genitourinary tract (e.g., renal,
  • 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) CrU 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
  • W 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 leukenuVlymphoma (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 leukenuVlymphoma
  • CTCL cutaneous T-cell lymphoma
  • LGF large granular lymphocytic leukemia
  • Hodgkin's disease Hodgkin's disease.
  • Combinations of Compounds described herein with other therapeutic agents it is desirable to treat a subject with a combination of agents, for example, if a patient is suffering from one conditions having multiple symptoms or is suffering from multiple conditions.
  • obesity is frequently associated with a plurality of inter-related diseases and conditions, such as, diabetes, cardiovascular disease, certain cancers, addictive behavior, and osteoarthritis.
  • Co-administration of two or more therapeutic compounds therefore could be used to treat obesity and one or more of the above listed associated diseases.
  • co-administration of drugs can achieve synergistic results, e.g., greater than additive results.
  • a combination (e.g., co-administration) of a plurality of therapeutic agents to treat a plurality of diseases or conditions simultaneously can improve the patient's quality of life by alleviating a plurality of symptoms, for example, symptoms associated with multiple diseases or conditions, and in some embodiments, allowing decreased dosing, which can reduce or eliminate undesirable side effects, due to drug synergism.
  • co-administration means that two or more agents are administered to a subject at the same time or within a time interval such that there is an overlap of an effect of each agent on the patient.
  • the agents can be administered simultaneously, for example, in a combined unit dose (providing simultaneous delivery of both agents).
  • the agents can be administered at a specified time interval, for example, an interval or minutes, hours, days, or weeks. In general, however, the agents are concurrently bioavailable, e.g., detectable in the subject.
  • a pharmaceutically effective dose of a ghrelin antagonist for example, a compound described herein, such as a compound of Formula I, II, III, IV, V, or VI, may be useful when administered (e.g., coadministered) as part of a combination therapy with an additional therapeutic agent.
  • Combinations of a ghrelin antagonist and a diabetic therapeutic agent A pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI may be useful when administered (e.g., coadministered) as part of a combination therapy with a second pharmaceutical agent for the simultaneous pharmacotherapy of diabetes and type II diabetes.
  • a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI may be useful when administered (e.g., coadministered) as part of a combination therapy with a second pharmaceutical agent for the simultaneous pharmacotherapy of diabetes and type II diabetes.
  • Exemplary anti-diabetics that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI include, for example, insulin preparations (e.g., short-acting insulins, such as, lispro insulin, insulin aspart, regular insulin, semilente insulin; intermediate-acting insulins, such as, NPH, lente insulin; and long-acting insulins, such as, PZl, ultrlente insulin, insulin glargine), and oral hypoglycemic agents (e.g., sulfonylureas (e.g., tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, glimepiride), meglitinides (e.g., repaglinide and nateglinide), ⁇ -gluco
  • a pharmaceutically effective dose of a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI maybe useful when administered (e.g., coadministered) as part of a combination therapy with metformin.
  • Cardiovascular disease refers to the group of diseases that affect the heart and/or blood vessels.
  • Exemplary CVDs include, for example, coronary heart disease, venous thromboembolic disease, endocarditis, congenital heart disease, myocardial ischemia, angina, arrhythmia, diseases of the aorta and its branches, diseases of the peripheral vascular system, orthostatic hypotension, hypertension, hyperlipidemia, arteriosclerosis, artherosclerosis, myocardial infarction, cardiac fibrillation, congestive heart failure, heart failure, and stroke.
  • a pharmaceutically effective dose of a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI maybe useful when administered (e.g., co-administered) as part of a combination therapy with one or more pharmaceutical agents directed to the treatment of CVD.
  • a plurality of pharmaceutical agents are available for the treatment of CVD, for example, cardiac glycosides (cardiotonics), agents for the treatment of myocardial ischemia, antiarrhythmic agents, antihypertensive agents, diuretics, antihyperlipidemic agents, anticoagulant, antiplatelet, thrombolytic agents, and antianemic agents.
  • cardiac glycosides cardiotonics
  • agents for the treatment of myocardial ischemia for example, antiarrhythmic agents, antihypertensive agents, diuretics, antihyperlipidemic agents, anticoagulant, antiplatelet, thrombolytic agents, and antianemic agents.
  • Myocardial ischemia or insufficient blood flow to part of the heart, can cause angina and heart attack.
  • Agents for the treatment of myocardial ischemia include antianginal agents and peripheral vasodilators.
  • Antianginal agents that may be administered (e.g., coadministered) as apart of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, nitrites, such as amyl nitrite; nitrates, such as nitroglycerin and isosorbide; ⁇ -blockers, such as propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, carteolol, oxprenolol, penbutolol, levatol, medroxalol, bucindolol, levobunolol, metipranol
  • Cardiac dysrhythmia is a group of conditions in which cardiac contractility is faster (tachycardia) or slower (bradycardia) than normal. Fibrillation of the ventricles (ventricular fibrillation), which involves a breakdown in the hearts inherent coordination during contraction, is life threatening.
  • Antiarrhythmic agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, cinchona alkaloids (e.g., quinidine), amides (e.g., procainamide, flecainide, lidocaine, bupivicaine, ropivicaine, and dispyramide), xylyl derivatives (e.g., lidocaine, and mexiletine), quaternary ammonium salts (e.g., bretylium), diiodobenzyloxyethylamines (e.g., amiodarone), ⁇ -b lockers, calcium antagonists, and hydantoins (e.g., phenytoin).
  • cinchona alkaloids e.g., qui
  • antiarrhythmic agents include, for example, tocainide, encainide, phenytoin, flecainide, tocainide, propafenone, bretylium, sotalol, ibutilide, dofctilidc, bcpridil, and moricizinc, propanolol, csmolol, artilidc, clof ⁇ lium, azimilidc, dronedarone, ersentilide, ibutilide, tedisamil, valvetilide, verapamil, diltaizem, digitalis, adenosine, nickel chloride, procainaltide, lignocaine, and magnesium ions.
  • Persistent hypertension also known as high blood pressure, is one of the leading predispositions for stroke, heart attack, heart failure, and arterial aneurysm.
  • Antihypertensive agents are classified into seven functionally distinct groups, as follows: First, diuretics; second, vasodilators; third, peripheral symphatholytics; fourth, central ⁇ 2 - sympathomimetics; fifth, calcium channel blockers; sixth, angiotensin-converting enzyme (ACE) inhibitors and angiotensin II -receptor antagonists.
  • ACE angiotensin-converting enzyme
  • Exemplary antihypertensive agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, thiazide diuretics, such as, bendrofluazide, bendroflumethiazide, benzthiazide, benzylhydrochlorothiazide, chlorothiazide, chlorthalidone, cyclopenthiazide, diucardin, diuril, enduron, esidrix, exna, HCTZ, hydrochlorothiazide, hydrodiuril, hydroflumethiazide, bendroflumethiazide, hydromox, hygroton, indapamide, lozol, methylchlorthiazide, metolazone, mykrox, naqua, nature
  • Hyperlipidemia which includes hyperlipoproteinemia, hypercholesterolemia, and hypertriglyceridemia, is characterized by elevated serum lipids.
  • Lipids exist in the bloodstream (e.g., cholesterol, cholesterol esters, phospholipids, and triglycerides) as lipoproteins, which are associated with chylomicrons, very low-density lipoproteins (VLDL), intermediate-density lipoproteins (IDL), low-density lipoproteins (LDL), and high-density lipoproteins (HDL).
  • VLDL very low-density lipoproteins
  • IDL intermediate-density lipoproteins
  • LDL low-density lipoproteins
  • HDL high-density lipoproteins
  • Hyperlipidemia or more specifically, hypercholesterolemia, is diagnosed in patients with total serum cholesterol greater than 200 mg/dl, or LDL levels greater than 130 mg/dl. Hyperlipidemia causes hardening of arteries, or atheros
  • Antihyperlipidemic agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula 1, 2, 3, or 4, include, for example, nonabsorbable agents (cholestyramine chloride and colestipol hydrochloride) and absorbable agents, including, statins (e.g., livostatin, simvastatin, pravastatin, fluvastatin, atorvastatin (commonly referred to as lipitor), and cerivastatin), fibrates (nicotinic acid, bezaf ⁇ brate, clinofibrate, clofibrate, colestimide, fenofibrate, gemf ⁇ brizil, probucol, simfibrate), and fatty fish oils containing large amounts of eicosapentaenoic acid and docosahexaenoic acid.
  • Congestive heart failure results when the heart can no longer pump sufficient supplies of blood to the body's organs.
  • Other conditions that decrease the ejection fraction of the heart include, for example, heart failure, atrial fibrillation, atrial flutter, and paroxysmal atrial tachycardia.
  • Agents used for the treatment of these conditions include the category of agents known as the cardiac glycosides. Cardiac glycosides increase mycocardial contractility and efficiency to improve systemic circulation and renal perfusion.
  • Common cardiac glycosides that may be administered (e.g., coadministered) in combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, xanthine formulations (e.g., aminophylline, choline theophylline, diprophylline, proxphylline), catecholamine formulations (e.g., dopamine, dobutamine, docarpamine), denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, and colforsin daropate.
  • a ghrelin antagonist for example, a compound described herein, such as,
  • ghrelin antagonist for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI
  • anticoagulant or antiplatelet agent include, for example, heparin (e.g., heparin sodium and heparin potassium) and low-molecular weight heparin fragments (enoxaparin, dalteparin, tinzaparin, ardeparin), low-molecular weight heparinoids (danaparoid), lepirudin, coumarin derivatives (warfarin and dicumarol), and phenindione.
  • heparin e.g., heparin sodium and heparin potassium
  • low-molecular weight heparin fragments enoxaparin, dalteparin, tinzaparin, ardeparin
  • low-molecular weight heparinoids danaparoid
  • lepirudin coumarin derivatives (warfarin and dicumarol)
  • antiplatelet agents include, for example, cyclooxygenase inhibitors (e.g., aspirin and indomethacin), ticlopidine, cilostazol, clopidogrel, dipyridamole, GPIIb/IIIa receptor antagonists (e.g., tirofiban and eptif ⁇ batide), anagrelide, and the Fab fragments of human monoclonal antibody to the GPIIb/IIIa receptor (e.g., Abciximab).
  • cyclooxygenase inhibitors e.g., aspirin and indomethacin
  • ticlopidine e.g., aspirin and indomethacin
  • cilostazol e.g., cilostazol
  • clopidogrel dipyridamole
  • dipyridamole e.g., cilostazol
  • clopidogrel dipyridamole
  • Thrombolytic agents are used to dissolve clots and reopen arteries or veins as a treatment for heart attack, stroke, deep vein thrombosis, pulmonary embolism, or occlusion of a peripheral artery.
  • Thrombolytic agents that may be administered (e.g., coadministered) as a part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, tissue plasminogen activators (e.g., alteplase, reteplase, and tenecteplase), streptokinase, anistreplase, and urokinase.
  • tissue plasminogen activators e.g., alteplase, reteplase, and tenecteplase
  • streptokinase anistreplase
  • Cancer is a disease characterized by abnormal cell growth and the formation of tumors or neoplasms, which posses the potential to spread, or metastasize.
  • the risk factors for cancer are multifactorial, and involve both genetic and environmental factors.
  • Obesity is linked to several types of cancer, including, for example, breast cancer, colon cancer, endometrial cancer, kidney cancer, esophageal cancer, and prostate cancer.
  • a pharmaceutically effective dose of a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may be administered (e.g., coadministered) as part of a combination therapy with an anticancer therapeutic or regimen.
  • anticancer therapies include palliative, adjuvant, neoadjuvant, and salvage chemotherapy and therapy using biological agents.
  • chemotherapeutic agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, alkylating agents (e.g., altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, and thiotepa), antitumor antibiotics (e.g., bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitox
  • Exemplary biological agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, interferon- ⁇ 2a and 2b (Roferon-A and lntron A, respectively), interleukin-2 (Aldesleukin and Proleukin), interleukin-11 (Oprelvekin and Neumega), the growth factor filgrastim (Neupogen), Pegf ⁇ lgrastim (Neulasta), Sargramostim (Leukine), epoetin alfa (Epogen or Procrit), darbepoetin alfa (Aranesp), and monoclonal antibodies, such as, Alemtuzumab, Gemtuzumab ozogamicin, Ibritumomab tuixetan, Rituximab
  • a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, FV, V, or VI, may also be used as part of a combination therapy with herbal anticancer therapies (e.g., ubenc tea, hoxsey therapy, iscador, chaparral, and Pau D' Arco), adjunctive herbal therapies, or with oncolytic adenoviruses.
  • herbal anticancer therapies e.g., oothc tea, hoxsey therapy, iscador, chaparral, and Pau D' Arco
  • a pharmaceutically effective dose of a ghrelin antagonist for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may, therefore, be usefully administered (e.g., coadministered) as part of a combination therapy with a second pharmaceutical for the pharmacotherapy of and addictive behavior.
  • Smoking cessation therapeutics that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, nicotine replacement, benzodiazepines (e.g., diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, and xanax), propanolol, clonidine, bupropion hydrochloride (Zyban and Wellbutrin), and CHANTIX.
  • benzodiazepines e.g., diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, and xanax
  • propanolol clo
  • Gambling cessation therapeutics that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, naltrexone, selective serotonin-reuptake inhibitor (SSRI) agents (e.g., citalopram, fluoxetine, zimelidine, sertraline, paroxetine), or a combination of Naltrexone and said SSRI agents.
  • SSRI selective serotonin-reuptake inhibitor
  • Alcoholism therapeutics that may be used as part of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, disulfiram, naltrexone, nalmefene (REVEX), acamprosate (Campral), buspirone, ondansetron (Zofran), and SSRI agents (e.g., citalopram (Celexa), escitalopram (Lexapro), fluoxetine (Prozac), paroxetine (Paxil), and sertraline (Zoloft).
  • a ghrelin antagonist include, for example, disulfiram, naltrexone, nalmefene (REVEX), acamprosate (Campral), buspirone, ondansetron (Zofran), and SSRI agents (e.g., citalopram (Celexa), es
  • Osteoarthritis formally known as degenerative joint disease, is a common chronic condition characterized by cartilage degeneration.
  • the major clinical features of osteoarthritis include pain and stiffness, leading to disability.
  • Obesity is an important risk factor for osteoarthritis in most joints, and in particular, the knee. Furthermore, weight loss is demonstrated to alleviate the symptoms and progression of osteoarthritis.
  • a pharmaceutically effective dose of a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may, therefore, be useful when administered (e.g., coadministered) as part of a combination therapy with an anti-osteoarthritic agent toalleviate the symptoms of osteoarthritis.
  • Exemplary anti-osteoarthritic agents that may be used as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula 1, 11, 111, IV, V, or Vl, include analgesics, for example, acetaminophen (Tylenol), nonsteroidal anti-inflammatory drugs (NSAIDs), such as, nonacetylated salicylates (e.g., salsalate (Disalcid) and magnesium salicylate (Doan's)), aspirin, diclofenac (Voltaren), etodolac (Lodine), ibuprofen (Motrin and Advil), naproxen (Naprosyn and Aleve), nabumeton (Relafen), sulindac (Clinoril), tolmetin (Tolectin), and COX-2 inhibitors, such as, celecoxib (Celebrex), refecoxib (Vi
  • anti-osteoarthritic agents include, for example, orally dosed tramadol (Ultram) and opiate analgesics (e.g., codeine and oxycodone); topical analgesics (e.g., capsaicin); intra-articular injections of corticosteroids or hyaluronic acid derivatives; adjunctive treatment with glucosamine, chondroitin, or S-adenosyl- methionine.
  • tramadol Ultram
  • opiate analgesics e.g., codeine and oxycodone
  • topical analgesics e.g., capsaicin
  • intra-articular injections of corticosteroids or hyaluronic acid derivatives e.g., capsaicin
  • adjunctive treatment with glucosamine, chondroitin, or S-adenosyl- methionine e.g., glucosamine
  • a pharmaceutically effective dose of a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may be useful when administered (e.g., coadministered) as part of a combination therapy with an agent used for the treatment of gout, including, for example, allopurinol, vorinostat, Zolinza, SAHA, depsipeptide (Romidepsin), MS-275, MGCDO 103, PXDlOl, Baceca, Savicol, LBH589, PCI-24781 , and ITF2357.
  • an agent used for the treatment of gout including, for example, allopurinol, vorinostat, Zolinza, SAHA, depsipeptide (Romidepsin), MS-275, MGCDO 103, PXDlOl, Baceca, Savicol, LBH589, PCI-24781 , and I
  • Obesity is associated with numerous other diseases and conditions for which there is no recommended or approved pharmacotherapy, including, for example, sleep apnea and Pickwikian syndrome. Obesity is also associated with abdominal hernia, varicose veins, gall bladder disease, and liver malfunction. In these cases, the most desirable strategy is prophylaxis through weight loss.
  • a pharmaceutically effective dose of a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may be useful when administered (e.g., coadministered) as part of a combination therapy with a weight loss pharmacotherapy.
  • Exemplary weight loss promoting agents include, for example, dexfenfluramine (Redux), amphetamine, methamphetamine, phenmetrazine (Preludin), phendimetrazine (Plegine), diethylpropion (Tenuate), phentermine (Lonamin), sibutramine (Meredia), and orlistat (Xenical).
  • a pharmaceutically effective dose of a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may be useful when administered (e.g., coadministered) as part of a combination therapy with a second pharmaceutical agent derived from the following classes of pharmaceuticals compounds, including, for example, insulin preparations, oral hypoglycemic agents, peroxisome proliferator-activated receptor gamma (PP AR ⁇ ) agonists, nitrates, nitrites, ⁇ -blockers, calcium channel antagonists, peripheral vasodilators, cinchona alkaloids, amides, xylyl derivatives, quaternary ammonium salts, diidobenzyloxyethylamines, hydantoins, diuretics, vasodilators, peripheral sympatholytics, central ⁇ 2-sympathomimetics, angiotensin-converting
  • Insulin preparations that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, short-acting insulins, such as, lispro insulin, insulin aspart, regular insulin, semilente insulin; intermediate-acting insulins, such as, NPH, lente insulin; and long-acting insulins, such as, PZI, ultrlente insulin, and insulin glargine.
  • short-acting insulins such as, lispro insulin, insulin aspart, regular insulin, semilente insulin
  • intermediate-acting insulins such as, NPH, lente insulin
  • long-acting insulins such as, PZI, ultrlente insulin, and insulin glargine.
  • Oral hypoglycemic agents that may be administered in combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, FV, V, or VI, include, for example, sulfonylureas (e.g., tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, glimepiride), meglitinides (e.g., repaglinide, nateglinide, and mitiglinide), ⁇ -glucosidase inhibitors (acarbose and miglitol), and insulin sensitizers, such as, biguanides (e.g., metformin), and thiazolidinediones (e.g., pioglitazone and rosiglitazone).
  • sulfonylureas e.g., tolbutamide, acetohexamide, chlorpropamide
  • Peroxisome proliferator-activated receptor gamma (PPAR ⁇ ) agonists that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, thiazolidinediones (e.g., pioglitazone and rosiglitazone).
  • Nitrates that may be administered in a combination therapy with a ghrelin antagonist include, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, nitroglycerin (e.g., glyceryl trinitrate, Nitro- Bid, Nitro-Stat, Nitrol, Nitro-Dur), isorbide dinitrate (e.g., Isordil, Sorbitrate, Dilatrate- SR), isorbide-5-mononitrate (e.g., Imdur, ISMO), and erythrityl tetranitrate (e.g., Cardilate).
  • nitroglycerin e.g., glyceryl trinitrate, Nitro- Bid, Nitro-Stat, Nitrol, Nitro-Dur
  • isorbide dinitrate e.g., Isordil, Sorbitrate, Dilatrate- SR
  • Nitrites that may be administered in a combination therapy with a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, amyl nitrate.
  • ⁇ -blockcrs that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, carteolol, oxprenolol, penbutolol, levatol, medroxalol, bucindolol, levobunolol, metipranolol, bisoprolol, nebivolol, betaxolol, celiprolol, sotalol, and propafenone.
  • a compound described herein such as
  • Calcium channel antagonists that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, amlodipine, arandipine, bamidipine, bencyclane, benidipine, bepridil, cilnidipine, cinnarizine, clentiazem, diltiazem, efonidipine, elgodipine, etafenone, flunarizine, filodepine, galopamil, isradipine, lacidipine, lercanidipine, lidoflazine, lomerizine, manidipine, mibefradil, nicardipine, nifedipine, nilvadepine, nimodipine, nisoldipine, nitrendipine, penyl
  • Peripheral vasodilators that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, dipyridamole, nylidrin hydrochloride, and isoxsuprine hydrochloride.
  • Cinchona alkaloids that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, quinidine and quinine.
  • a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, quinidine and quinine.
  • Amides that may be administered in a combination therapy with a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, procainamide, flecainide, lidocaine, bupivicaine, ropivicaine, and dispyramide.
  • Xylyl derivatives that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI include, for example, lidocaine and mexiletine.
  • Diidobenzyloxyethylamines that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, amiodarone.
  • Hydantoins that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, phenytoin.
  • Diuretics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, thiazide diuretics, such as, bendrofluazide, bendroflumethiazide, benzthiazide, benzylhydrochlorothiazide, chlorothiazide, chlorthalidone, cyclopenthiazide, diucardin, diuril, enduron, esidrix, exna, HCTZ, hydrochlorothiazide, hydrodiuril, hydroflumethiazide, bendroflumethiazide, hydromox, hygroton, indapamide, lozol, methylchlorthiazide, metolazone, mykrox, naqua, naturetin, oretic, penfiutizide, polythiazide, qui
  • Vasodilators that may be administered in a combination therapy with a ghrelin antagonist, for example, a compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, bencyclane, cinnarizine, citicoline, cyclandate, cyclonicate, diazoxide, ebumamonine, flunarizine, hydralazine, ibudilast, ifenprodil, lomerizine, minoxidil, naphlole, nikamate, nimodipine, nitroprusside, nofedoline, nosergoline, papaverine, pentifylline, pentoxifylline, phenoxezyl, prostacyclin derivatives, vichizyl, vincamin, and vinpocetine.
  • a compounds described herein such as, a compound of Formula I, II, III, IV, V, or VI
  • Peripheral sympatholytics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, acebutol, atenolol, betaxolol, bisoprolol, carteolol, carvediolol, doxazosin, guanadrel, guanethidine, labetalol, metoprolol, nadolol, penbutolol, pindolol, prazosin, propanolol, reserpine, terazosin, and timolol.
  • Central ⁇ -sympathomimetics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, clom ' dine, guanabenz, guanfacine, and methyldopa.
  • Angiotensin-converting enzyme (ACE) inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, alacepril, benazepril, captopril, ceranopril, cilazopril, delapril, enalapril, fosinopril, imidapril, lisinopril, mobertpril, moexipril, pentopril, perindopril, quinapril, ramipril, randolapril, spirapril, temocapril, trandolapril, and zofenopril.
  • alacepril for example, alacepril, benazepril, captopril, ceranopril, cilazopril, delapril, enala
  • Angiotensin II-receptor antagonists that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, candesartan, eprosartan, ibersartan, losartan, telmisartan, and valsartan.
  • HMG CoA reductase inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, livostatin, simvastatin, pravastatin, fluvastatin, atorvastatin (commonly referred to as lipitor), and cerivastatin.
  • Fibrates that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, nicotinic acid, bezafibrate, clinofibrate, clofibrate, colestimide, fenofibrate, gemfibrizil, probucol, and simfibrate.
  • a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, nicotinic acid, bezafibrate, clinofibrate, clofibrate, colestimide, fenofibrate, gemfibrizil, probucol, and simfibrate.
  • Antiarrhythmic agents that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, tocainide, encainide, phenytoin, flecainide, tocainide, propafenone, bretylium, sotalol, ibutilide, dofetilide, bepridil, and moricizine, propanolol, esmolol, artilide, clofilium, azimilide, dronedarone, ersentilide, ibutilide, tedisamil, verapamil, diltaizem, digitalis, adenosine, nickel chloride, procainaltide, lignocaine, and magnesium ions.
  • a compound described herein such as, a compound of Formula I, II, III, IV, V, or VI
  • Cardiac glycosides that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, xanthine formulations (e.g., aminophylline, choline theophylline, diprophylline, proxphylline), catecholamine formulations (e.g., dopamine, dobutamine, docarpamine), denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, and colforsin daropate.
  • a compound described herein such as, a compound of Formula I, II, III, IV, V, or VI
  • Heparin agents that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, heparin sodium and heparin potassium, low- molecular weight heparin fragments (enoxaparin, dalteparin, tinzaparin, ardeparin), and low-molecular weight heparinoids (danaparoid).
  • a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI
  • heparin sodium and heparin potassium include, for example, heparin sodium and heparin potassium, low- molecular weight heparin fragments (enoxaparin, dalteparin, tinzaparin, ardeparin), and low-molecular weight heparinoids (danaparoid).
  • Coumarin derivatives that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, warfarin and dicumarol.
  • Histone deacetylase (HDAC) inhibitors may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as a compound of Formula I, II, III, IV, V, or VI.
  • HDAC inhibitors such as HDAC I/II inhibitors
  • small molecular weight carboxylates e.g., less than about 250 amu
  • hydroxamic acids e.g., benzamides, epoxyketones, cyclic peptides, and hybrid molecules.
  • HDAC inhibitors such as HDAC I/II inhibitors
  • small molecular weight carboxylates e.g., less than about 250 amu
  • hydroxamic acids e.g., less than about 250 amu
  • benzamides e.g., less than about 250 amu
  • hydroxamic acids e.g., less than about 250 amu
  • benzamides e.g., less than about 250 amu
  • hydroxamic acids e.g., less than about 250 amu
  • benzamides e.g., less than about 250 amu
  • hydroxamic acids e.g., less than about 250 amu
  • benzamides e.g., less
  • Non-limiting examples of negative regulators of type I/II HDACs include: Suberoylanilide Hydroxamic Acid (SAHA (e.g., MK0683, vorinostat) and other hydroxamic acids), Suberoyl bis- hydroxamic Acid, BML-210, Depudecin (e.g., (-)-Depudecin), HC Toxin, Nullscript (4- ( 1 ,3 -Dioxo- 1 H,3H-benzo[de]isoquinolin-2-yl)-N-hydroxybutanamide), Phenylbutyrate (e.g., sodium phenylbutyrate) and Valproic Acid (and other short chain fatty acids), Scriptaid, Suramin Sodium, Trichostatin A (TSA), APHA Compound 8, Apicidin, Sodium Butyrate, pivaloyloxymethyl butyrate (Pivanex, AN-9), Trapoxin B,
  • SAHA Suberoylanilide Hydro
  • CI- 994 i.e., N-acetyl dinaline
  • MS-27-275 MS-27-275
  • MGCD0103 i.e., NVP-LAQ-824
  • inhibitors include, for example, dominant negative forms of the HDACs (e.g., catalytically inactive forms) siRNA inhibitors of the HDACs, and antibodies that specifically bind to the HDACs.
  • Inhibitors are available, e.g., from BIOMOL International, Fukasawa, Merck Biosciences, Novartis, Gloucester Pharmaceuticals, Aton Pharma, Titan Pharmaceuticals, Schering AG, Pharmion, MethylGene, and Sigma Aldrich.
  • Sirtuin SIRT inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, 2-anilinobenzamides (nicotinamide, carba-NAD + , EX-527, sirtinol, para-sirtinol, JFD00244, splitomicin, HR73, cambinol) and indoles (Napper A., et al. (2005). J. Med.
  • 2-anilinobenzamides nicotinamide, carba-NAD + , EX-527, sirtinol, para-sirtinol, JFD00244, splitomicin, HR73, cambinol
  • indoles Napper A., et al. (2005). J. Med.
  • Xanthine oxidase inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, Allopurinol.
  • Alkylating agents that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula 1, H, 111, IV, V, or VI, include, for example, altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, and thiotepa.
  • Antitumor antibiotics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitoxantrone, and valrubicin.
  • Antimetabolites that may be administered in a combination therapy with a ghrelin antagonist include, for example, capecitabine, cladribine, cytarabine, fludarabine, floxuridine, 5-fluorouracil, gemicabine, hydroxyurea, 6-mercaptopurine, methotrexate, pentostatin, and 6-thioguanine.
  • Mitotic inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, docetaxel, paclitaxel, vinblastine, vincristine, and vinorelbine.
  • Topoisomerase inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, etoposide, irinotecan, teniposide, and topotecan.
  • Enzymes that may be administered in a combination therapy with a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, asparaginase and pagasparaginase.
  • Protein tyrosine kinase inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, FV, V, or VI, include, for example, imatinib mesylate.
  • Hormonal agents that may be administered in a combination therapy with a ghrelin antagonist include, for example, a compound described herein, such as, a compound of Formula 1, II, III, IV, V, or Vl, include, for example, adrenocorticoids (e.g., dexamethasone, methylprednisolone, and prednisone), estrogens (e.g., diethylstilbestrol and estradiol), progestins (e.g., medroxyprogesterone and megesterol acetate), antiestrogens (e.g., fulvestrant, tamoxifen, and toremifene), androgens (e.g., testosterone, methyltestosterone, and fiuoxymesterone) and antiandrogens (e.g., bicalutamide, flutamide, and nilutamide).
  • adrenocorticoids e.g., dexa
  • Aromatase inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, aminoglutethimide, anastrozole, exemestane, and letrozole.
  • LHRH analogues that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, leuprolide and goserelin.
  • a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, leuprolide and goserelin.
  • Biological agents that may be administered in a combination therapy with a ghrelin antagonist include, for example, interferon- ⁇ 2a and 2b (Roferon-A and Intron A, respectively), interleukin-2 (Aldesleukin and Proleukin), interleukin-11 (Oprelvekin and Neumega), the growth factor filgrastim (Neupogen), Pegfilgrastim (Neulasta), Sargramostim (Leukine), epoetin alfa (Epogen or Procrit), darbepoetin alfa (Aranesp), and monoclonal antibodies, such as, Alemtuzumab, Gemtuzumab ozogamicin, Ibritumomab tuixetan, Rituximab, and Trastuzumab.
  • interferon- ⁇ 2a and 2b Rosferon-A and Intron A, respectively
  • interleukin-2 Aldesleukin and Proleukin
  • a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may also be administered in a combination therapy with an herbal therapy (e.g., ubenc tea, hoxsey therapy, iscador, chaparral, and Pau D' Arco) or with an oncolytic adenovirus.
  • an herbal therapy e.g., ubenc tea, hoxsey therapy, iscador, chaparral, and Pau D' Arco
  • Benzodiazepines that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, and xanax.
  • Opioid-receptor antagonists that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, Ul, IV, V, or VI, include, for example, tramadol and naltrexone.
  • Antidepressants that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, selective serotonin reuptake inhibitors (SSRIs) (e.g., citalopram (Celexa), escitalopram (Lexapro), fluoxetine (Prozac), paroxetine (Paxil), sertraline (Zoloft), serotonin and norepinephrine reuptake inhibitors (SNRIs) (e.g., duloxetine (Cymbalta) and venlafaxine (Effexor), norepinephrine and dopamine reuptake inhibitors (NDRIs) (e.g., Wellbutrin and Zyban), tetracyclic antidepressants (e.g., mirtazapine (Remeron), combined reuptake inhibitors and receptor
  • Analgesics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, paracetamol (acetaminophen and indomethacin), non-steroidal anti-inflammatory drugs (NSAIDs) (aspirin, ibuprofen, salsalate, and magnesium salicylate), and cyclooxygenase-2 (COX-2) inhibitors (e.g., celecoxib (Celebrex), refecoxib (Vioxx), and valdecoxib (Bextra)), opiates (e.g., codeine, oxycodone, hydrocodone, diamorphine, pethidine, tramadol, and bupreno ⁇ hine), corticosteroids (e.g., prednisone, prednisolone, cortisone, and hydrocortisone), and
  • Amphetamines and sympathomimetic amines that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, dextroamphetamine, benzedrine, ritalin, phenmetrazine (Preludin), phendimetrazine (Plegine), methamphetamine, diethylpropion (Tenuate), phentermine (Lonamin), sibrutamine (Meredia), and orlistat (Xenical).
  • a ghrelin antagonist for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, dextroamphetamine, benzedrine, ritalin, phenmetrazine (Preludin), phendimetrazine (Plegine), methamp
  • 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 treat one or more of the following disorders: cachexia, wasting, stimulating growth hormone release in elderly humans, patients with cancer, heart failure, or AIDS; treating growth hormone deficient adults; prevention of cataboHc 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
  • 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.
  • 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.
  • 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.
  • 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.
  • the informational material e.g., instructions
  • the informational material is provided in printed matter, e.g., a printed text, drawing, and/or photograph, e.g., a label or printed sheet.
  • the informational material can also be provided in other formats, such as computer readable material, video recording, or audio recording.
  • 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 an compound described herein and/or its use in the methods described herein.
  • the informational material can also be provided in any combination of formats.
  • 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.
  • the other ingredients can be included in the kit, but in different compositions or containers than the compound described herein.
  • 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.
  • the liquid solution preferably is an aqueous solution, with a sterile aqueous solution being preferred.
  • a suitable 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.
  • the kit contains separate containers, dividers or compartments for the composition and informational material.
  • the composition can be contained in a bottle, vial, or syringe, and the informational material can be contained in a plastic sleeve or packet.
  • the separate elements of the kit are contained within a single, undivided container.
  • the composition is contained in a bottle, vial or syringe that has attached thereto the informational material in the form of a label.
  • 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.
  • 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.
  • 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.
  • the device is an implantable delivery device.
  • Example 2 Synthesis of (R)-3-(benzyloxy)-2-(3- (diethylamino * )propylsiilfonamido)propanoic acid
  • Thionyl chloride 50ml, 685mmol
  • N,N-3-diethylammopropane sulfonic acid hydrochloride (2) was added to N,N-3-diethylammopropane sulfonic acid hydrochloride (2) and heated to reflux whilst stirring for 8hours.
  • the solution was allowed to cool to ambient temperatures and then evaporated in vacuo to give N,N-3-diethylaminopropanesulfonyl chloride hyrochloride (3) as a grey/white, very hydroscopic solid (7.9g, 92%).
  • the product was stored under a nitrogen atmosphere.
  • Example 3 Synthests of (R)-3-(diethylamino)-N-(l-(3-subsituted-l,2,4-oxadiazol-5- vI)-3-phenyIpropyl)propane-l-suIfonamide and (R)-2-(diethyIamino)-N-(l-(3- substituted-l,2,4-oxadiazol-5-yl)-3-phenylpropyl)ethanesulfonamide R ⁇ N 1
  • N-BOC-Homophenylalanine 200mg, 0.72mmol was dissolved in EtOAc (10ml) and cooled to O 0 C. To this solution was added CDI (128mg, 0.79mmol) and stirred at O 0 C for 15mins, and then a further 2hrs whilst warming to ambient temperatures. The amidoxime (2, 0.79mmol) was then added and the reaction mixture was stirred overnight. The reaction mixture was then washed with sat. ⁇ aHC ⁇ 3( aq ) (10ml) then brine (10ml) before drying OVCrNa 2 SO 4 , filtering and evaporating to dryness in vacuo.
  • N,N-Diethylamine (2ml) was added to (6) and stirred at ambient temperatures overnight. This was then evaporated to dryness in vacuo.
  • the crude product was purified by silica gel column chromatography, ehiting with a mixture of MeOH/DCM (typically 5/95).
  • Boc-HomoPhe-methylester (5) (3g, 10.2mmol) was taken in hydrazine hydrate (99% solution (10ml) at room temperature and continued the reaction at 80 oC for 3 h.
  • the reaction mixture was diluted with dichloromethane (20ml), washed with saturated sodium bicarbonate solution, water followed by brine solution. The organic layer was dried over sodium sulfate, filtered, concentrated under reduced pressure to provide the crude material. The crude residue was purified by flash column chromatography on silica gel using methanol: dichloromethane (4:96) as eluent. The obtained compound was taken in dichloromethane and washed with water.
  • Example 6 Reduced ghrelin-induced food intake in mice dosed ip with compound
  • Example 7 Exemplary compounds were orally bioavailable
  • Example 8 28 days oral dosing of compounds 57 and 94 caused a decrease in body weight with no apparent effect on food intake
  • mice received high fat diet and water ad lib to establish diet-induced obesity, and for the duration of the experiment. After acclimating to individual housing for at least 5 days, mice were then acclimated to bid oral gavage with vehicle for 7 days. Once acclimated, mice were culled into equal groups (n>10/group) according to body weight and compound dosing began. Mice received either vehicle or compound (bid, po) for periods up to 28 days, during which body weight and food intake were measured daily. Representative cohorts (based on BW) for each group were removed at 7, 14, 21 or 28 days for glucose tolerance tests and tissue analysis. As shown in Figure 3a and Figure 3b, administration of compound 57 and compound 94 provided a decrease in body weight. No apparent effect on food intake was observed.
  • Example 9 7 to 28 days oral dosing of compounds 57 and 95 improved insulin sensitivity
  • Figure 4b shows the data demonstrating improved insulin sensitivity of mice dosed with compound 57 at 7 days
  • Figure 4e shows the data demonstrating improved insulin sensitivity of mice dosed with compound 94 at 7 days
  • Figure 4c shows the data demonstrating improved insulin sensitivity of mice dosed with compound 57 at 21 days
  • Figure 4f shows the data demonstrating improved insulin sensitivity of mice dosed with compound 94 at 28 days.
  • ExamplelO 28 day dosing of compound 57 and compound 94 results in improved plasma HbAIc and cholesterol levels
  • mice are sacrificed by CO 2 asphyxiation and terminal blood samples were collected via cardiac puncture.
  • HbAIc whole blood is analyzed using a DCA2000+ glycated hemoglobin analyzer (Bayer Healthcare, Indianapolis, IN). Triglycerides, total cholesterol, and HDL-cholesterol were all measured using a Cholestech LDX lipid profile analyzer (Cholestech, Hayward, CA). As shown in Figure 6 and Figure 7, both plasma
  • HbAIc and cholesterol levels were improved at 28 days in animals treated with compound 57 and compound 94 versus animals treated with vehicle alone.
  • mice that had been acclimated to individual housing and oral gavage dosing were fasted for 16 hours overnight. The following morning a blood sample was removed from the tail for baseline fasted blood glucose measurements. This process was repeated after 14 days oral bid dosing with compound 57. Whereas fasted blood glucose levels continued to rise in vehicle treated animals, there was no change in mice treated with the ghrelin antagonist. Thus, compound treatment blocked the development of hyperglycemia in this model of type 2 diabetes.
  • Figure 7 illustrates the improvement in fasting blood glucose levels of mice treated with compound 57.

Abstract

Compounds that modulate GHS-R are described, for examples compounds formula (I).

Description

Ghrelin modulating compounds and combinations thereof
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 oxadiazole and triazole 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.
The compounds, including stereoisomers thereof, can be created either singly, in small clusters, or in a combinatorial fashion to give structurally diverse libraries of compounds.
In one aspect, the invention features a compound of formula (I)
Figure imgf000002_0001
formula (I) wherein,
R1 is hydrogen, halo (e.g., fluoro), aryl, heteroaryl, arylalkyl, heteroarylalkyl, cyclyl, cyclylalkyl, heterocyclyl, heterocyclylalkyl, alkyl, alkenyl, alkynyl, or R1 can be taken together with R2 or R3 to form a ring; each of which is optionally substituted with 1-4 R6; k' is a bond, O, C(O), C(O)O, OC(O), C(O)NR3, NR3C(O), S, SO, SO2, CR2=CR2, or C≡C; n is 0-6, preferably 1-3;
R2 is hydrogen, halo (e.g., fluoro), Ci-C6 alkyl, C2-C<5 alkenyl, or C2-C6 alkynyl; or R2 can be taken together with R1 to form a ring;
R3 is hydrogen, Ci-Cβ alkyl, C2-Cg alkenyl, or C2-Ce alkynyl, or R3 can be taken together with R2, R4, or R5 to form a ring; each of which can be optionally substituted with 1-2 R6';
A is
R7a R7a
-ζ-(CH2)x C (CH2)y-|- -|— N-(CH2), C (CH2)y-<- R7b , R8 R7b
Figure imgf000003_0001
x and y are each independently 0-6;
M is aryl, heteroaryl, cyclyl, or heterocyclyl, each of which is optionally substituted with 1-4 R9; R4 and R3 arc each independently hydrogen, alkyl, alkenyl, haloalkyl, cyclyl, or heterocyclyl, or R4 and R1 can be taken together to form a heterocyclic ring, or R4 and R5 can be taken together to form an azido moiety, or one or both of R4 and R5 can independently be joined to one or both of R7a and R7b to form one or more bridges between the nitrogen to which the R4 and R5 are attached and R7a and R7b, wherein each bridge contains 1 to 5 carbons; or one or both of R4 and R5 can independently be joined to one or both of R7a and R7b to form to form one or more heterocyclic rings including the nitrogen to which the R4 and R5 are attached, or one or both of R4 and R5 can independently be joined to R3 to form a ring, or one or both of R4 and R5 can independently be joined to R8 to form a ring; wherein each R4 and R5 are optionally independently substituted with 1-5 halo, 1-3 hydroxy, 1-3 alkyl, 1-3 alkoxy, 1-3 oxo, 1-3 amino, 1-3 alkylamino, 1-3 dialklyamino, 1-3 nitrile, or 1-3 haloalkyl;
Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R10; each R6 and R6 are independently halo, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl, heteroaryl, alkoxy, haloalkyl, haloalkyloxy, haloalkylthio, acetyl, cyano, nitro, hydroxy, oxo, C(O)OR2, OC(O)R2, N(R3)2, C(O)N(R3)2, NR3C(O)R2, or
SR2
R7a and R7b are each independently hydrogen, alkyl, alkenyl, haloalkyl, cyclyl, cyclylalkyl, or heterocyclyl; or one or both of R7a and R7b can independently be joined to one or both of R4 and R5 to form one or more bridges between the nitrogen to which the R4 and R5 are attached and R7a and R7b, wherein each bridge contains 1 to 5 carbons; or one or both of R7a and R7b can independently be joined to one or both of R4 and R5 to form to form one or more heterocyclic rings including the nitrogen to which the R4 and R5 are attached, or one or both of R7a and R7*5 can independently be joined with R8 to form a ring; wherein each R7a and R7b can be independently optionally substituted with 1- 5 halo, 1-3 hydroxy, 1-3 alkyl, 1-3 alkoxy, 1-3 amino, 1-3 alkylamino, 1-3 dialklyamino, 1-3 nitrile, or 1-3 haloalkyl;
R8 is hydrogen or Ci-C6 alkyl, or R8 can be joined with R4, R5, R7a or R7b to form a ring;
R9 is halo, alkyl, cyclyl, heterocyclyl, aryl, heteroaryl, alkoxy, haloalkyl, haloalkyloxy, haloalkylthio, acetyl, cyano, nitro, hydroxy, oxo, C(O)OR2, OC(O)R2, N(R2)2, C(O)N(R2)2, NR2C(O)R2, SR2; each R10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR11, - NR11R11', -CF3, -SOR12, -SO2R12, -OC(O)R11, -SO2NR12R12', -(CH2)mR14 or R15; each of which is optionally independently substituted with 1-3 R16;
R11 and R11' are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
R12 and R12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R12 and R12 taken together can be cyclized to form -(CH2)qX(CH2)s-; wherein each R12 and R12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR11, alkoxy, heterocycloalkyl, -NR11C(O)NR11R11', -C(O)NR11R11', -NR11C(O)R11', -CN, oxo, -NR11SO2R11', -OC(O)R11, -SO2NR11R11', -SOR13, -S(O)2R13, -COOH and -C(O)OR13; each R13 is independently alkyl, aryl, arylalkyl, heteroaryl, or heteroarylalkyl, each of which may optionally be substituted with -(CH2)WOH; each R14 is independently alkoxy, alkoxycarbonyl, -C(O)NR12R12', -NR11R11', - C(O)R12, -NR11C(O)NR11R11' or -N-heteroaryl; each R15 is independently -(CH2)PN(R12)C(O)R12', -(CH2)PCN, - (CH2)pN(R12)C(O)OR12', -(CH2)pN(R12)C(O)NR12R12', -(CH2)PN(R12)SO2R12, -
(CH2)PSO2NR12R12', -(CH2)pC(O)NR12R12', -(CH2)PC(O)OR12, -(CH2)POC(O)OR12, - (CH2)POC(O)R12, -(CH2)POC(O)NR12R12', -(CH2)pN(R12)SO2NR12R12', -(CH2)POR12, - (CH2)pOC(O)N(R12)(CH2)mOH, -(CH2)PSOR12, -(CH2)PSO2R12, -(CH2)PNR11R11 or - (CH2)pOCH2C(O)N(R12)(CH2)mOH; each R16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, -
(CH2)PNR11C(O)NR11R11', -(CH2)PC(O)NR11R11', -(CH2)PNR11C(O)R11', -CN, - (CH2)PNR11SO2R11', -(CH2)POC(O)R1 \ -(CH2)PSO2NR11R111, -(CH2)PSOR13, - (CH2)pC00H or -(CH2)PC(O)OR13;
X is CR11R11', O, S, S(O), S(O)2, or NR11; m is an integer between 1 and 6; p is an integer from O to 5; q and s are each independently an integer between 1 and 3; and w is an integer between 0 and 5.
In some embodiments, formula (I), comprises an enriched preparation of formula
(T)
Figure imgf000006_0001
formula (Y).
In some embodiments, formula (I), comprises an enriched preparation of formula (I")
Figure imgf000006_0002
formula (I").
In some embodiments, n is 1; k' is a bond or O; and
R1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl. In some embodiments, n is 1; k' is O; and R1 is arylalkyl.
For example, R1 can be phenylmethyl. In some embodiments, n is 2; k' is a bond; and R1 is aryl.
In some embodiments, n is O or 1; k' is a bond; and R1 is alkyl, for example unsubstituted or substituted with one R6. For example, R1 can be a branched alkyl such as one of the following .
Figure imgf000007_0001
In some embodiments, R2 is hydrogen or C1-C3 alkyl. In some embodiments n is 0 and k' is a bond. Exemplary R1 moieties include methyl, and ethyl. Preferred R1 moieties include methyl. In some embodiments R1 is unsubstituted methyl or methyl or ethyl substituted with C(O)N(R3)2.
In some embodiments n is 0 and k' is a bond, and R1 and R2 are both methyl.
In some embodiments, n is 0; k' is a bond; and
R1 is hydrogen.
In some embodiments R3 is hydrogen.
In some embodiments, R and R together form a heterocyclic ring such as a pyrrolidine or an azetidine ring. The heterocyclic ring can be unsubstituted or substituted, for example, with 1-2 R6.
In some embodiments, R1 and R2 together form a ring.
In some embodiments, A is
R7a R7a
-p"(CH2)x C (CH2)y-5- -<-N— (CH2)X C (CH2)yγ
R7b or R8 R7b
For example, A can be
Figure imgf000007_0002
or A can be
Figure imgf000008_0001
; wherein
R7a and R7b are H; x is 1 ; and y is 0 or 1. In some embodiments,
A is CH2CH2 or CH2CH2CH2; and each R4 and R5 is independently alkyl, or R4 and R5, when taken together, form a heterocyclic ring. In some embodiments, R'a and R can each be H.
In some embodiments, at least one of R7a or R7b is taken together with at least one of R4 or R5 to form a heterocyclic ring including the nitrogen to which the R4 and R5 are attached.
In some embodiments, R7a and R7b are each independently alkyl; R4 and R5 are each independently hydrogen or alkyl; and x and y are each independently 0 or 1. In some embodiments,
Figure imgf000008_0002
, taken together is
Figure imgf000008_0003
In some embodiments,
Figure imgf000009_0001
taken together is
Figure imgf000009_0002
In some embodiments,
Figure imgf000009_0003
taken together is
Figure imgf000009_0004
In some embodiments,
Figure imgf000009_0005
taken together is
Figure imgf000009_0006
In some embodiments,
Figure imgf000009_0007
, taken together is
Figure imgf000009_0008
In some embodiments, Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
In some embodiments, Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
In some embodiments Y is substituted with one R10. R10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon. In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroary alkyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety. The cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy. In some embodiments Y is oxadiazole or triazole. In another aspect, the invention features a compound of formula (II),
Figure imgf000010_0001
formula (II) wherein,
Q1 J Q2, Q3 and Q4 together with the carbon to which they are attached form a heteroaryl moiety, and each Q1, Q2, Q3 and Q4 is independently S, O, N, CR2, CR10, NR2, or NR 10
In some embodiments, the compound of formula (II), comprises an enriched preparation of formula (II')
Figure imgf000011_0001
formula (H'). In some embodiments, the compound of formula (II), comprises an enriched preparation of formula (H")
Figure imgf000011_0002
formula (H").
In some embodiments, Q1 and Q4 are each independently S, O, N, or NR10. In some embodiments, Q1 and Q3 are each independently S, O, N, or NR10.
In some embodiments, Q2 is CR2 or CR10.
In some embodiments, Q2 is S, O, N, or NR10.
In some embodiments, at least one of Q2 or Q3 is CR2 or CR10.
In some embodiments, at least two of Q1, Q2, Q3, or Q4 is S, O, N, or NR10. In some embodiments, Q1, Q2, and Q3 are each independently S, O, N, or NR10.
In some embodiments, Q1 is NR10.
In some embodiments, one of Q2, Q3, or Q4 is CR2'
In some embodiments, Q2 is CR10.
In some embodiments, Q3 is CR2' In some embodiments, Q1, Q2, Q3 and Q4 together form
Figure imgf000012_0001
In some embodiments, Q1 is NR2.
In some embodiments, Q , Q , Q and Q together form
Figure imgf000012_0002
In some embodiments, Q1 is NR10. In another aspect, the invention features a compound of formula (III),
Figure imgf000012_0003
wherein, Z , Z , Z , Z , and Z together form an aryl or heteroaryl moiety, and each Z , Z ,
Z3, Z4, and Z5 is independently N, CR10, or CR2.
In some embodiments, the compound of formula (III), comprises an enriched preparation of formula (HF)
Figure imgf000012_0004
formula (IIP).
In some embodiments, the compound of formula (III), comprises an enriched preparation of formula (HF)
Figure imgf000012_0005
formula (IIP). In some embodiments, one of Z1, Z2, Z3, Z4, and Z5 is N.
In some embodiments, two of Z1, Z2, Z3, Z4, and Z5 are N.
In some embodiments, three of Z1, Z2, Z3, Z4, and Z5 is N.
In some embodiments, two of Z1 and Z2 are N. In some embodiments, two of Z1 and Z3 are N.
In some embodiments, two of Z1 and Z4 are N.
In some embodiments, two of Z1, Z3, and Z5 are N.
In some embodiments, the compound is a compound of formula (I), wherein Y is substituted with a single substituent R10. For example, R10 can be aryl or heteroaryl, optionally substituted with up to three independent R16.
In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy. In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety. The cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is Rπ.
In some embodiments, Y is substituted with a second R10, for example an alkyl, halo or alkoxy.
In some embodiments, R1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl; k' is a bond or O; n is 1 or 2;
R2 and R3 are both hydrogen;
A is
Figure imgf000014_0001
x and y are each independently 0-6; R4 and R5 are each independently hydrogen or alkyl;
Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R10; each R10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR11, - NR11R11', -CF3, -SOR12, -SO2R12, -OC(O)R11, -SO2NR12R12', -(CH2)mR14 or R15; each of which is optionally independently substituted with 1-3 R16; R11 and R11 are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
R12 and R12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R12 and R12 taken together can be cyclized to form -(CH2)qX(CH2)s-; wherein each R12 and R12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR11, alkoxy, heterocycloalkyl, -NR11C(O)NR11R11', -C(O)NR11R11', -NR11C(O)R11', -CN, oxo, -NR11SO2R11', -OC(O)R11, -SO2NR11R11', -SOR13, -S(O)2R13, -COOH and -C(O)OR13; each R13 is independently alkyl, aryl, arylalkyl, heteroaryl, or heteroarylalkyl, each of which may optionally be substituted with -(CH2)w0H; each R14 is independently alkoxy, alkoxycarbonyl, -C(O)NR12R12', -NR11R11', - C(O)R12, -NR11C(O)NR11R11' or -N-heteroaryl; each R15 is independently -(CH2)PN(R12)C(O)R12', -(CH2)PCN, - (CH2)PN(R12)C(O)OR12', -(CH2)pN(R12)C(O)NR12R12', -(CH2)PN(R12)SO2R12, - (CH2)pSO2NR12R12', -(CH2)pC(O)NR12R12', -(CH2)PC(O)OR12, -(CH2)POC(O)OR12, - (CH2)POC(O)R12, -(CH2)POC(O)NR12R12', -(CH2)PN(R12)SO2NR12R12', -(CH2)POR12, - (CH2)pOC(O)N(R12XCH2)mOH, -(CH2)PSOR12, -(CH2)PSO2R12,
Figure imgf000015_0001
or - (CH2)pOCH2C(O)N(R12)(CH2)mOH; each R16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH2)PNR11C(O)NR1 1R11', -(CH2)PC(O)NR11R11', -(CH2)PNR11C(O)R1'', -CN, - (CH2)PNR11SO2R11', -(CH2)POC(O)R1 \ -(CH2)PSO2NR11R11', -(CH2)PSOR13, - (CH2)pC00H or -(CH2)PC(O)OR13;
X is CR11R11', O, S, S(O), S(O)2, or NR11; m is an integer between 1 and 6; p is an integer from O to 5; q and s are each independently an integer between 1 and 3; and w is an integer between O and 5.
For example, in some embodiments, n is 1; k' is a bond or O; and R1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl. In some embodiments, n is 1 ; k' is O; and R1 is arylalkyl, for example phenylmethyl. In some embodiments, n is 2; k'is a bond; and R1 is aryl. For example, in some embodiments, R7a and R7b are H; x is 1 ; and y is O or 1. In some embodiments, A is CH2CH2 or CH2CH2CH2.
In some embodiments, each R4 and R5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
In some embodiments, Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
In some embodiments, Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms. In some embodiments Y is substituted with one R10. R10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon.
In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroary alkyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety. The cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments Y is oxadiazole or triazole.
In some embodiments, Y is "Q , wherein Ql is O or NR2, preferably O or NH. In some embodiments, R10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R16. In some embodiments, R10 is substituted with one R16, such as halo (e.g., fluoro or chloro) or alkoxy. In some embodiments, the compounds has a formula (Ia)
Figure imgf000016_0001
formula (Ia)
In some embodiments, R1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl; k' is a bond or O; n is 1 or 2; A is CH2, CH2CH2, or CH2CH2CH2;
R4 and R^ are each independently hydrogen or alkyl;
Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R10; each R10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR11, - NR11R11', -CF3, -SOR12, -SO2R12, -OC(O)R11, -SO2NR12R12', -(CH2)mR14 or R15; each of which is optionally independently substituted with 1-3 R16;
R11 and R11 are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
R12 and R12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R12 and R12 taken together can be cyclized to form -(CH2)qX(CH2)s-; wherein each R12 and R12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR11, alkoxy, heterocycloalkyl, -NR11C(O)NR11R11', -C(O)NR11R11', -NR11C(O)R11', -CN, oxo, -NR11SO2R11', -OC(O)R11, -SO2NR11R11', -SOR13, -S(O)2R13, -COOH and -C(O)OR13; each R13 is independently alkyl, aryl, arylalkyl, heteroaryl, or heteroarylalkyl, each of which may optionally be substituted with -(CH2)WOH; each R14 is independently alkoxy, alkoxycarbonyl, -C(O)NR12R12', -NR11R11', -
C(O)R12, -NR11C(O)NR11R11' or -N-heteroaryl; each R15 is independently -(CH2)PN(R12)C(O)R12', -(CH2)PCN, - (CH2)pN(R12)C(O)OR12', -(CH2)PN(R12)C(O)NR12R12', -(CH2)PN(R12)SO2R12, - (CH2)PSO2NR12R12', -(CH2)pC(O)NR12R12', -(CH2)PC(O)OR12, -(CH2)POC(O)OR12, - (CH2)pOC(O)R12, -(CH2)POC(O)NR12R12', -(CH2)PN(R12)SO2NR12R12', -(CH2)POR12, - (CH2)pOC(O)N(Ri2)(CH2)mOH, -(CH2)PSOR12, -(CH2)PSO2R12, -(CH2)PNRURU or - (CH2)pOCH2C(O)N(R12)(CH2)mOH; each R16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH2)PNR11C(O)NR11R11', -(CH2)PC(O)NR11R11', -(CH^NR11C(O)R11', -CN, - (CH2)pNRnSO2Rir, -(CH2)POC(O)R1 \ -(CH2)PSO2NR11R11', -(CH2)PSOR13, - (CH2)PC00H or -(CH2)PC(O)OR13;
X is CR11R11', O, S, S(O), S(O)2, or NR11; m is an integer between 1 and 6; p is an integer from O to 5; q and s are each independently an integer between 1 and 3; and w is an integer between O and 5.
For example, in some embodiments, n is 1; k' is a bond or O; and R1 is aryl, heteroaryl, arylalkyl, or heteroarylatkyl. In some embodiments, n is 1; k' is O; and R1 is arylalkyl, for example phenylmethyl. In some embodiments, n is 2; k'is a bond; and R1 is aryl.
In some embodiments, A is CH2CH2 or CH2CH2CH2, preferably CH2CH2CH2.
In some embodiments, each R4 and R5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
In some embodiments, Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
In some embodiments, Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms. In some embodiments Y is substituted with one R10. R10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon.
In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy. In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy. In some embodiments, R10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety. The cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments Y is oxadiazole or triazole.
Figure imgf000019_0001
In some embodiments, Y is ""Q , wherein Ql is O or NR2, preferably O
NH. In some embodiments, R10 is aryl, arylalkyl, heteroaryl, or hctcroarylalkyl, for example optionally substituted with one or more R16. In some embodiments, R10 is substituted with one R16, such as halo (e.g., fluoro or chloro) or alkoxy. In some embodiments, R1 is hydrogen or alkyl, for example unsubstituted or substituted with one R6; n is 0 or 1 ; k' is a bond; and
R2 and R3 each independently hydrogen or Ci-C6 alkyl;
A is
Figure imgf000019_0002
x and y arc each independently 0-6; R4 and R5 are each independently hydrogen or alkyl; Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R10; each R10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR11, -
NR11R11', -CF3, -SOR12, -SO2R12, -OC(O)R11, -SO2NR12R12', -(CH2)JR14 or R15; each of which is optionally independently substituted with 1-3 R16;
R1 ] and R1 r are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl; R12 and R12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R12 and R12 taken together can be cyclized to form -(CH2)qX(CH2)s-; wherein each R12 and R12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR11, alkoxy, heterocycloalkyl, -NR11C(O)NR11R11', -C(O)NR11R11', -NR11C(O)R11', -CN, oxo, -NR11SO2R11', -OC(O)R11, -SO2NR11R11', -SOR13, -S(O)2R13, -COOH and -C(O)OR13; each Rn is independently alkyl, aryl, arylalkyl, heteroaryl, or heteroarylalkyl, each of which may optionally be substituted with -(CH2)WOH; each R14 is independently alkoxy, alkoxycarbonyl, -C(O)NR12R12', -NR11R11', - C(O)R12, -NR11C(O)NR11R11' or -N-heteroaryl; each R15 is independently -(CH2)PN(R12)C(O)R12', -(CH2)PCN, - (CH2)PN(R12)C(O)OR12', -(CH2)PN(R12)C(O)NR12R12', -(CH2)PN(R12)SO2R12, - (CH2)PSO2NR12R12', -(CH2)PC(O)NR12R12', -(CH2)PC(O)OR12, -(CH2)POC(O)OR12, - (CH2)POC(O)R12, -(CH2)POC(O)NR12R12', -(CH2)pN(R12)SO2NR12R12', -(CH2)POR12, - (CH2)pOC(O)N(R12)(CH2)mOH, -(CH2)PSOR12, -(CH2)PSO2R12, -(CH^pNR11R11 or - (CH2)pOCH2C(O)N(R12)(CH2)mOH; each R16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH2)PNR11C(O)NR11R11', -(CH2)PC(O)NR11R11', -(CH2)PNR11C(O)R11', -CN, - (CH2)PNR11SO2R11', -(CH2)POC(O)R1 \ -(CH2)PSO2NR11R11', -(CH2)PSOR13, - (CH2)pC00H or -(CH2)PC(O)OR13;
X is CR11R11', O, S, S(O), S(O)2, or NR11; m is an integer between 1 and 6; p is an integer from O to 5; q and s are each independently an integer between 1 and 3; and w is an integer between 0 and 5. In some embodiments, n is 0 or 1; k' is a bond; and
R1 is alkyl, for example unsubstituted or substituted with one R6. In some embodiments n is 0 and k' is a bond. Exemplary R1 moieties include methyl, and ethyl. Preferred R1 moieties include methyl, In some embodiments R1 is unsubstituted methyl or methyl or ethyl substituted with C(O)N (R3)2. In some embodiments, n is 0 or 1; k' is a bond; and R1 is alkyl, for example unsubstituted or substituted with one R6. For example, R1 can be a branched alkyl such as one of the following .
Figure imgf000021_0001
In some embodiments n is 0 and k' is a bond, and R1 and R3 are both methyl.
In some embodiments, n is 0; k' is a bond; and
R1 is hydrogen.
In some embodiments, A is CH2CH2 or CH2CH2CH2, preferably CH2CH2CH2.
In some embodiments, each R4 and R5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
In some embodiments, Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety. In some embodiments, Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
In some embodiments Y is substituted with one R10. R10 can be positioned, for example, 1 ,3 relative to the point of attachment of Y to the adjacent chain carbon or 1 ,2 relative to the point of attachment of Y to the adjacent chain carbon.
In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy. In some embodiments, R10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety. The cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments Y is oxadiazole or triazole.
In some embodiments, Y is
Figure imgf000022_0001
> wherein Ql is O or NR2, preferably O or
NH. In some embodiments, R10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R16. In some embodiments, R10 is substituted with one R16, such as halo (e.g., fluoro or chloro) or alkoxy. In some embodiments, the compounds has a formula (Ib)
Figure imgf000023_0001
In some embodiments, R1 is hydrogen or alkyl; A is CH2, CH2CH2, or CH2CH2CH2; R2 is hydrogen or C1-C3 alkyl;
R4 and R5 are each independently hydrogen or alkyl;
Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R10; each R10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR11, - NR11R11', -CF3, -SOR12, -SO2R12, -OC(O)R11, -SO2NR12R12', -(CH2)mR14 or R15; each of which is optionally independently substituted with 1-3 R16;
R11 and R11 are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
R12 and R are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R12 and R12 taken together can be cyclized to form -(CH2)qX(CH2)s-; wherein each R12 and R12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR11, alkoxy, heterocycloalkyl, -NR11C(O)NR11R11', -C(O)NR11R11', -NR11C(O)R11', -CN, oxo, -NR11SO2R11', -OC(O)R11, -SO2NR11R11', -SOR13, -S(O)2R13, -COOH and -C(O)OR13; each R13 is independently alkyl, aryl, arylalkyl, heteroaryl, or heteroarylalkyl, each of which may optionally be substituted with -(CH2)WOH; each R14 is independently alkoxy, alkoxycarbonyl, -C(O)NR12R12', -NR11R11', -
C(O)R12, -NR11C(O)NR11R11' or -N-heteroaryl; each R15 is independently -(CH2)PN(R12)C(O)R12', -(CH2)PCN, - (CH2)pN(R12)C(O)OR12', -(CH2)pN(R12)C(O)NR12R12', -(CH2)PN(R12)SO2R12, - (CH2)PSO2NR12R12', -(CH2)PC(O)NR12R12', -(CH2)PC(O)OR12, -(CH2)POC(O)OR12, - (CH2)pOC(O)R12, -(CH2)POC(O)NR12R12', -(CH2)PN(R12)SO2NR12R12', -(CH2)POR12, - (CH2)pOC(O)N(R12)(CH2)mOH, -(CH2)PSOR12, -(CH2)PSO2R12, -(CH2)pNRnRn or - (CH2)pOCH2C(O)N(R12)(CH2)mOH; each R1 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH2)PNR11C(O)NR11R11', -(CH2)PC(O)NR11R11', -(CH2)PNR11C(O)R11', -CN, - (CH2)pNRnSO2Rir, -(CH2)POC(O)RU, -(CH2)PSO2NR11R11', -(CH2)PSOR13, - (CH2)pC00H or -(CH2)PC(O)OR13;
X is CR11R11 ', O, S, S(O), S(O)2, or NR11; m is an integer between 1 and 6; p is an integer from O to 5; q and s are each independently an integer between 1 and 3; and w is an integer between 0 and 5.
In some embodiments, A is CH2CH2 or CH2CH2CH2, preferably CH2CH2CH2.
In some embodiments, each R4 and R5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
In some embodiments, Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
In some embodiments, Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
In some embodiments Y is substituted with one R10. R10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon. In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety. The cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments Y is oxadiazole or triazole.
In some embodiments, Y is
Figure imgf000025_0001
NH. In some embodiments, R10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R16. In some embodiments, R10 is substituted with one R16, such as halo (e.g., fluoro or chloro) or alkoxy.
In some embodiments, R1 and R3 together form a heterocyclic ring such as a pyrrolidine or an azetidine ring (The heterocyclic ring can be unsubstituted or substituted, for example, with 1-2 R6.); n is 0 or 1 ; k' is a bond;
R2 hydrogen or C1-Ce alkyl, preferably hydrogen;
A is
Figure imgf000025_0002
x and y are each independently 0-6;
R4 and R^ arc each independently hydrogen or alkyl; Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R10; each R10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR11, -
NR11R11', -CF3, -SOR12, -SO2R12, -OC(O)R11, -SO2NR12R12', -(CH2)mR14 or R15; each of which is optionally independently substituted with 1-3 R16;
R1 ' and R1 ' are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl; R12 and R12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R12 and R12 taken together can be cyclized to form -(CH2)qX(CH2)s-; wherein each R12 and R12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR11, alkoxy, heterocycloalkyl, -NR11C(O)NR11R11', -C(O)NR11R11', -NR11C(O)R11', -CN, oxo, -NR11SO2R11', -OC(O)R11, -SO2NR11R11', -SOR13, -S(O)2R13, -COOH and -C(O)OR13; each R13 is independently alkyl, aryl, arylalkyl, heteroaryl, or heteroarylalkyl, each of which may optionally be substituted with -(CH2)WOH; each R14 is independently alkoxy, alkoxycarbonyl, -C(O)NR12R12', -NR11R11', - C(O)R12, -NR11C(O)NR11R11' or -N-heteroaryl; each R15 is independently -(CH2)PN(R12)C(O)R12', -(CH2)PCN, - (CH2)pN(R12)C(O)OR12', -(CH2)PN(R12)C(O)NR12R12', -(CH2)PN(R12)SO2R12, - (CH2)PSO2NR12R12', -(CH2)PC(O)NR12R12', -(CH2)PC(O)OR12, -(CH2)POC(O)OR12, - (CH2)POC(O)R12, -(CH2)POC(O)NR12R12', -(CH2)pN(R12)SO2NR12R12', -(CH2)POR12, - (CH2)pOC(O)N(R12)(CH2)mOH, -(CH2)PSOR12, -(CH2)PSO2R12, -(CH2)PNR11R11 or - (CH2)pOCH2C(O)N(R12)(CH2)mOH; each R16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH2)PNR11C(O)NR11R11', -(CH2)PC(O)NR11R11', -(CH2)PNR11C(O)R11', -CN, - (CH2)PNR11SO2R11', -(CH2)POC(O)R1 \ -(CH2)PSO2NR11R11', -(CH2)PSOR13, - (CH2)pC00H or -(CH2)PC(O)OR13;
X is CR11R11', O, S, S(O), S(O)2, or NR11; m is an integer between 1 and 6; p is an integer from O to 5; q and s are each independently an integer between 1 and 3; and w is an integer between 0 and 5. In some embodiments, A is CH2CH2 or CH2CH2CH2, preferably CH2CH2CH2.
In some embodiments, each R4 and R5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
In some embodiments, Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
In some embodiments, Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroaryalkyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety. The cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments Y is oxadiazole or triazole.
Figure imgf000028_0001
In some embodiments, Y is "Q , wherein Ql is O or NR2, preferably O NH. In some embodiments, R10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R16. In some embodiments, R10 is substituted with one R16, such as halo (e.g., fluoro or chloro) or alkoxy. In some embodiments, the compounds has a formula (Ic) ψ] R A
-L-N-SO2 A— (
& R5 formula (Ic) n is O, 1, 2, 3, or 4; preferably 1 or 2;
A is CH2, CH2CH2, or CH2CH2CH2; R4 and R5 are each independently hydrogen or alkyl;
Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with l-4 R10; each R10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR11, -
NR11R11', -CF3, -SOR12, -SO2R12, -OC(O)R11, -SO2NR12R12', -(CH2)mR14 or R15; each of which is optionally independently substituted with 1-3 R16;
R11 and R11 are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl; R12 and R12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R12 and R12' taken together can be cyclized to form -(CH2)qX(CH2)s-; wherein each R12 and R12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR11, alkoxy, heterocycloalkyl, -NR11C(O)NR11R11', -C(O)NR11R11', -NR11C(O)R11', -CN, oxo, -NR11SO2R11', -OC(O)R11, -SO2NR11R11', -SOR13, -S(O)2R13, -COOH and -C(O)OR13; each R13 is independently alkyl, aryl, arylalkyl, heteroaryl, or heteroarylalkyl, each of which may optionally be substituted with -(CH2)WOH; each R14 is independently alkoxy, alkoxycarbonyl, -C(O)NR12R12', -NR11R11', - C(O)R12, -NR11C(O)NR11R11' or -N-heteroaryl; each R15 is independently -(CH2)PN(R12)C(O)R12', -(CH2)PCN, - (CH2)pN(R12)C(O)OR12', -(CH2)PN(R12)C(O)NR12R12', -(CH2)PN(R12)SO2R12, - (CH2)PSO2NR12R12', -(CH2)pC(O)NR12R12', -(CH2)PC(O)OR12, -(CH2)POC(O)OR12, - (CH2)POC(O)R12, -(CH2)POC(O)NR12R12', -(CH2)PN(R12)SO2NR12R12', -(CH2)POR12, -
(CH2)pOC(O)N(R12)(CH2)mOH, -(CH2)PSOR12, -(CH2)PSO2R12, -(CH2)PNR11R11 or - (CH2)POCH2C(O)N(R12)(CH2)mOH; each R16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH2)PNR11C(O)NR11R11', -(CH2)PC(O)NR11R11', -(CH2)PNR11C(O)R11', -CN, - (CH2)PNR11SO2R11', -(CH2)POC(O)R1 \ -(CH2)PSO2NR11R11', -(CH2)PSOR13, - (CH2)pC00H or -(CH2)PC(O)OR13;
X is CR11R11', O, S, S(O), S(O)2, or NR11; m is an integer between 1 and 6; p is an integer from O to 5; q and s are each independently an integer between 1 and 3; and w is an integer between O and 5.
In some embodiments, A is CH2CH2 or CH2CH2CH2, preferably CH2CH2CH2.
In some embodiments, each R4 and R5 is independently alkyl, for example, methyl or ethyl, preferably ethyl.
In some embodiments, Y is a monocyclic heteroaromatic moiety, for example a nitrogen containing heteraromatic moiety such as a nitrogen containing five membered heteraromatic moiety.
In some embodiments, Y is a heterocyclic moiety containing at least two heteroatoms, for example, a five membered heterocyclic moiety containing at least two heteroatoms or at least three heteroatoms.
In some embodiments Y is substituted with one R10. R10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or 1,2 relative to the point of attachment of Y to the adjacent chain carbon. In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, oxazolyl, thiazolyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, benzimidazolyl, benzoxazolyl, benzofuranyl, benzothiophenyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 is arylalkyl or heteroarylalky, for example a monocyclic or bicyclic arylalkyl or monocyclic or bicyclic heteroary alkyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments, R10 includes an unsaturated or partially unsaturated cyclic moiety, for example a cyclyl or heterocyclyl moiety. The cyclic moiety can either be directly attached to Y or attached via a linker such as an alkylenyl linker. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, cyano, or methoxy.
In some embodiments Y is oxadiazole or triazole.
R10
K V-N T V|-
In some embodiments, Y is "Q , wherein Ql is O or NR2, preferably O or
NH. In some embodiments, R10 is aryl, arylalkyl, heteroaryl, or heteroarylalkyl, for example optionally substituted with one or more R16. In some embodiments, R10 is substituted with one R16, such as halo (e.g., fluoro or chloro) or alkoxy.
In another aspect, the invention features a compound of formula (IV)
Figure imgf000030_0001
formula (IV) wherein,
R1 is hydrogen, aryl, heteroaryl, arylalkyl, heteroarylalkyl, cyclyl, cyclylalkyl, heterocyclyl, heterocyclylalkyl, alkyl, alkenyl, alkynyl, or R1 can be taken together with R2 or R3 to form a ring; each of which is optionally substituted with 1-4 R6; k' is a bond, O, C(O), C(O)O, OC(O), C(O)NR3, NR3C(O), S, SO, SO2, CR2=CR2, or C≡C; n is 0-6, preferably 1-3;
R2 is hydrogen, CI-CO alkyl, C2-Cβ alkenyl, or C2-CO alkynyl; A' is heterocyclyl; optionally substituted with 1-3 R9;
Y is a monocyclic aryl or monocyclic heteroaryl; each of which is optionally substituted with 1-4 R10; each R6 is independently halo, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl, heteroaryl, alkoxy, haloalkyl, haloalkyloxy, haloalkylthio, acetyl, cyano, nitro, hydroxy, oxo, C(O)OR2, OC(O)R2, N(R3)2, C(O)N(R3)2, NR3C(O)R2, or SR2;
R9 is halo, alkyl, cyclyl, heterocyclyl, aryl, heteroaryl, alkoxy, haloalkyl, haloalkyloxy, haloalkylthio, acetyl, cyano, nitro, hydroxy, oxo, C(O)OR2, OC(O)R2, N(R2)2, C(O)N(R2)2, NR2C(O)R2, SR2; each R10 is independently alkyl, alkenyl, alkynyl, halo, cyano, carbonyl, aryl, arylalkyl, arylalkenyl, arylalkynyl, cyclyl, cyclylalkyl, alkoxy, alkoxyalkyl, aryloxy, aryloxyalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl, -OR11, - NR11R11', -CF3, -SO2R12, -OC(O)R11, -SO2NR12R12', -(CH2)mR14 or R15; each of which is optionally independently substituted with 1-3 R16;
R11 and R11 are each independently hydrogen, alkyl, alkenyl, alkynyl, cyclyl, heterocyclyl, aryl or heteroaryl;
R12 and R12 are each independently hydrogen, alkyl, alkenyl, alkynyl, alkylthioalkyl, alkoxyalkyl, aryl, arylalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, heterocycloalkyl or cyclyl, cyclylalkyl, or R12 and R12 taken together can be cyclized to form -(CH2)qX(CH2)sS wherein each R12 and R12 may independently optionally be substituted with 1 to 3 substituents selected from the group consisting of halogen, OR11, alkoxy, heterocycloalkyl, -NR11C(O)NR11R11', -C(O)NR11R11', -NR11C(O)R11', -CN, oxo, -NR11SO2R11', -OC(O)R11, -SO2NR11R11', -SOR13, -S(O)2R13, -COOH and -C(O)OR13; each R13 is independently alkyl, aryl, arylalkyl, heteroaryl, or heteroarylalkyl, each of which may optionally be substituted with -(CH2)WOH; each R14 is independently alkoxy, alkoxycarbonyl, -C(O)NR12R12', -NR11R11', -
C(O)R12, -NR11C(O)NR11R11' or -N-heteroaryl; each R15 is independently heterocycloalkyl, heteroaryl, -CN, - (CH2)pN(R12)C(O)R12', -(CH2)pCN, -(CH2)PN(R12)C(O)OR12', - (CH2)pN(R12)C(O)NR12R12', -(CH2)PN(R12)SO2R12, -(CH2)PSO2NR12R12', - (CH2)PC(O)NR12R12', -(CH2)PC(O)OR12, -(CH2)POC(O)OR12, -(CH2)POC(O)R12, - (CH2)POC(O)NR12R12', -(CH2)PN(R12)SO2NR12R12', -(CH2)POR12, -
(CH2)pOC(O)N(R12)(CH2)mOH, -(CH2)PSOR12 or -(CH2)pOCH2C(O)N(R12)(CH2)mOH; each R16 is independently halo, alkyl, alkenyl, alkynyl, alkoxy, - (CH2)PNR11C(O)NR1 1R1 r, -(CH2)pC(0)NRnRn', -(CH2)PNR11C(O)R11', -CN, - (CH2)PNR11SO2R11', -(CH2)POC(O)R1 \ -(CH2)PSO2NR11R11', -(CH2)PSOR13, - (CH2)pC00H or -(CH2)PC(O)OR13;
X is CR11R11', O, S, S(O), S(O)2, or NR11; m is an integer between 1 and 6; p is an integer from O to 5. q and s are each independently an integer between 1 and 3; and w is an integer between 0 and 5.
In some embodiments, the compound of formula (IV), comprises an enriched preparation of formula (IV)
R1
Figure imgf000032_0001
formula (IV).
In some embodiments, the compound of formula (IV), comprises an enriched preparation of formula (IV")
Figure imgf000032_0002
formula (IV").
In some embodiments, A' is a 5 or 6 membered heterocyclyl. In some embodiments, the 5 or 6 membered heterocyclyl includes at least two nitrogen atoms.
In some embodiments, A' is
Figure imgf000033_0001
In some embodiments, A' is substituted with one R9, for example, N(R2)2.
In some embodiments, n is 1; k' is a bond or O; and
R1 is aryl, heteroaryl, arylalkyl, or heteroarylalkyl. In some embodiments, n is 1; k' is O; and
R1 is arylalkyl.
For example, R1 can be phenylmethyl. In some embodiments, n is 2; k' is a bond; and
R1 is aryl.
In some embodiments, Y is a monocyclic heteroaromatic moiety, for example, a nitrogen containing heteraromatic moiety, such as a nitrogen containing 5 membered heteraromatic moiety.
In some embodiments, Y is a heterocyclic moiety containing at least two heteroatoms, for example, a 5 membered heterocyclic moiety containing at least two heteroatoms or a heterocyclic moiety containing at least 3 heteroatoms. In some embodiments, Y is substituted with 1 R10. The R10 can be positioned, for example, 1,3 relative to the point of attachment of Y to the adjacent chain carbon or can be positioned, for example, 1,2 relative to the point of attachment of Y to the adjacent chain carbon. In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, or methoxy. In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, imidazolyl, benzoxazolyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, or methoxy.
In some embodiments, Y is oxadiazole or triazole. In another aspect, the invention features a compound of formula (V),
Figure imgf000034_0001
formula (V) wherein,
Q1, Q2, Q3 and Q4 together with the carbon to which they are attached form a heteroaryl moiety, and each Q1, Q2, Q3 and Q4 is independently S, O, N, CR2, CR10, NR2, or NR10.
In some embodiments, the compound of formula (V), comprises an enriched preparation of formula (V)
Figure imgf000034_0002
formula (V).
In some embodiments, the compound of formula (V), comprises an enriched preparation of formula (V")
Figure imgf000035_0001
formula (V").
In some embodiments, Q1 and Q4 are each independently S, O, N, or NR10. In some embodiments, Q1 and Q3 are each independently S, O, N, or NR10.
In some embodiments, Q2 is CR2 or CR10.
In some embodiments, Q2 is S, O, N, or NR10.
In some embodiments, at least one of Q2 or Q3 is CR2 or CR10.
In some embodiments, at least two of Q1, Q2, Q3, or Q4 is S, O, N, or NR10. In some embodiments, Q1, Q2, and Q3 are each independently S, O, N, or NR10.
In some embodiments, Q1 is NR10.
In some embodiments, one of Q2, Q3, or Q4 is CR2'
In some embodiments, Q2 is CR10.
In some embodiments, Q3 is CR2' In some embodiments, Q1, Q2, Q3 and Q4 together form
Figure imgf000035_0002
In some embodiments, Q1 is NR2.
In some embodiments, Q1, Q2, Q3 and Q4 together form
Figure imgf000035_0003
In some embodiments, Q1 is NR10.
In another aspect, the invention features a compound of formula (Vl),
Figure imgf000035_0004
formula (VI) wherein
Z1, Z2, Z3, Z4, and Z5 together form an aryl or heteroaryl moiety, and each Z1, Z2, Z3, and Z5 is independently N, CR10, or CR2.
In some embodiments, the compound of formula (VI), comprises an enriched preparation of a compound of formula (VF).
R1
Figure imgf000036_0001
formula (VI').
In some embodiments, the compound of formula (VI), comprises an enriched preparation of a compound of formula (VI").
R1 z< z^1 TJ.
Z3 V t— c C-- NNN M----SSaSOUυU2222---V Z2 Z Z11 R2 '
formula (VI").
In some embodiments, one of Z1, Z2, Z3, Z4, and Z5 is N. In some embodiments, two of Z1, Z2, Z3, Z4, and Z5 are N.
In some embodiments, three of Z1, Z2, Z3, Z4, and Z5 is N. In some embodiments, two of Z1 and Z2 are N. In some embodiments, two of Z1 and Z3 are N. In some embodiments, two of Z1 and Z4 are N. In some embodiments, two of Z1, Z3, and Z5 are N.
In some embodiments, the compound is a compound of formula (IV), wherein Y is substituted with a single substituent R10. For example, R10 can be aryl or heteroaryl, optionally substituted with up to three independent R16.
In some embodiments, R10 is aryl or heteroaryl, for example a monocyclic aryl or monocyclic heteroaryl such as phenyl, pyridyl, or thiophenyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, or methoxy.
In some embodiments, R10 is a bicyclic heteroaryl, for example indolyl, imidazolyl, benzoxazolyl, or benzthiazolyl. In some embodiments, R10 is substituted with 1-3 R16. In some embodiments, R16 is halo, alkyl, or alkoxy, for example chloro, fluoro, methyl, or methoxy.
In some embodiments, R10 is R15.
In some embodiments, Y is substituted with a second R10, for example an alkyl, halo or alkoxy. 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 administred 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 another aspect, the invention features a compound listed in Table 1.
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), (II), (III), (IV), (V), or (VI). 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 stereoconfϊgurations 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 diastereromers of the 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), (II), (III), (IV), (V), or (VI). 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%. Diastereromer, 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.
In one aspect the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent. In one embodiment, the additional therapeutic agent is an insulin preparation, an oral hypoglycemic, a peroxisome proliferator-activated receptor gamma agonist, a nitrate, a nitrite, a β-blocker, a calcium channel antagonist, a peripheral vasodilator, a cinchona alkaloid, an amide, a quaternary ammonium salt, a diidobenzyloxyethylamine, a hydantoin, a diuretic, a vasodilator, a peripheral sympatholytic, a central α2 -sympathomimetic, an angiotensin-converting enzyme inhibitor, an angiotensin II-receptor antagonist, a HMG CoA reductase inhibitor, a fibrate, an antiarrhythmic, a cardiac glycoside, a heparin, a coumarin derivative, a histone deacetylase (HDAC) inhibitor, a sirtuin (SIRT) inhibitor, a xanthine oxidase inhibitor, an alkylating agent, an antitumor antibiotic, an antimetabolite, a mitotic inhibitor, a topoisomerase inhibitor, an enzyme, a protein tyrosine kinase inhibitor, a hormonal agent, an aromatase inhibitor, a LHRH analogue, a biological agent, a herbal remedy, an oncolytic adenovirus, a benzodiazepan, an opioid-receptor antagonist, an antidepressant, an analgesic, a hyaluronic acid derivative, or an amphetamine.
In one aspect, the invention features a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI, and an additional therapeutic agent. In one embodiment, the additional therapeutic agent is an insulin preparation, an oral hypoglycemic, a peroxisome proliferator-activated receptor gamma agonist, a nitrate, a nitrite, a β-blocker, a calcium channel antagonist, a peripheral vasodilator, a cinchona alkaloid, an amide, a quaternary ammonium salt, a diidobenzyloxyethylamine, a hydantoin, a diuretic, a vasodilator, a peripheral sympatholytic, a central α2 -sympathomimetic, an angiotensin-converting enzyme inhibitor, an angiotensin II-receptor antagonist, a HMG CoA reductase inhibitor, a fibrate, an antiarrhythmic, a cardiac glycoside, a heparin, a coumarin derivative, a histone deacetylase (HDAC) inhibitor, a sirtuin (SIRT) inhibitor, a xanthine oxidase inhibitor, an alkylating agent, an antitumor antibiotic, an antimetabolite, a mitotic inhibitor, a topoisomerase inhibitor, an enzyme, a protein tyrosine kinase inhibitor, a hormonal agent, an aromatase inhibitor, a LHRH analogue, a biological agent, a herbal remedy, an oncolytic adenovirus, a benzodiazepan, an opioid-receptor antagonist, an antidepressant, an analgesic, a hyaluronic acid derivative, or an amphetamine. In another embodiment, the invention includes a tablet or capsule. In a further embodiment, the tablet or capsule will feature an enteric coating.
In one aspect, the invention features a method of treating a disorder in a subject. The method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent. In one embodiment, the ghrelin antagonist is co-administered with the additional therapeutic agent. In an embodiment, the ghrelin antagonist and the additional therapeutic agent are administered in a single dosage form. In an embodiment, the ghrelin antagonist and the additional therapeutic agent are provided in amounts so as to provide a synergistic effect. In yet a further embodiment, the ghrelin antagonist and the additional therapeutic agent are administered one time daily. In an embodiment, the ghrelin antagonist and the additional therapeutic agent are administered twice daily. In an embodiment, the ghrelin antagonist and the additional therapeutic agent are administered three times daily. In an embodiment, the ghrelin antagonist and the additional therapeutic agent are administered within 1 hour before or two hours after a meal. In an embodiment, the ghrelin antagonist and the additional therapeutic agent are administered without regard to food. In an embodiment, the disorder is obesity. In an alternative embodiment, the disorder is a combination of disorders. In a further alternative embodiment, the combination of disorders is obesity and an additional disorder. In yet a further alternative embodiment, the additional therapeutic agent is an insulin preparation, an oral hypoglycemic, a peroxisome proliferator-activated receptor gamma agonist, a nitrate, a nitrite, a β- blocker, a calcium channel antagonist, a peripheral vasodilator, a cinchona alkaloid, an amide, a quaternary ammonium salt, a diidobenzyloxyethylamine, a hydantoin, a diuretic, a vasodilator, a peripheral sympatholytic, a central α2-sympathomimctic, an angiotcnsin- converting enzyme inhibitor, an angiotensin ϊϊ-receρtor antagonist, a HMG CoA reductase inhibitor, a fϊbrate, an antiarrhythmic, a cardiac glycoside, a heparin, a coumarin derivative, a histone deacetylase (HDAC) inhibitor, a sirtuin (SIRT) inhibitor, a xanthine oxidase inhibitor, an alkylating agent, an antitumor antibiotic, an antimetabolite, a mitotic inhibitor, a topoisomerase inhibitor, an enzyme, a protein tyrosine kinase inhibitor, a hormonal agent, an aromatase inhibitor, a LHRH analogue, a biological agent, a herbal remedy, an oncolytic adenovirus, a benzodiazepan, an opioid-receptor antagonist, an antidepressant, an analgesic, a hyaluronic acid derivative, or an amphetamine. In an embodiment, the insulin preparation is a short acting insulin, regular insulin, semilente insulin, intermediate acting insulin, lente insulin, or long acting insulin. In an embodiment, the oral hypoglycemic is a sulfonylurea, a mcglitinidc, α-glucosidasc inhibitor, or an insulin sensitizer. Tn an embodiment, the sulfonylurea is tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, or glimepiride. In an embodiment, the meglitinide is mitiglinide, repaglinide or nateglinide. In an embodiment, the α- glucosidase inhibitor is acarbose, miglitol, voglibose (N-l(l,3-dihydroxy-2-propyl) valiolamine, and N-substituted derivatives thereof, and N-substituted pseudo-amino sugars. In an embodiment, the insulin sensitizer is a biguanide, or a thiazolidinedione. In an embodiment, the biguanide is metformin. In an embodiment, the thiazolidinedione is pioglitazone or rosiglitazone. In an embodiment, the peroxisome proliferator-activated receptor gamma agonist is a thiazolidinedione. In an embodiment, the nitrate is nitroglycerin, isorbide, isorbide-5-mononitrate, or erythrityl tetranitrate. In an embodiment, the nitrite is amyl nitrate. In an embodiment, the β-blocker is propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, carteolol, oxprenolol, penbutolol, levatol, medroxalol, bucindolol, levobunolol, metipranolol, bisoprolol, nebivolol, betaxolol, celiprolol, sotalol, or propafenone. In an embodiment, the calcium channel antagonist is amlodipine, arandipine, bamidipine, bencyclane, benidipine, bepridil, cilnidipine, cinnarizine, clentiazem, diltiazem, efonidipine, elgodipine, etafenone, flunarizine, filodepine, galopamil, isradipine, lacidipine, lercanidipine, lidoflazine, lomerizine, manidipine, mibefradil, nicardipine, nifedipine, nilvadepine, nimodipine, nisoldipine, nitrendipine, penylamine, perhexiline, phendilin, semotiadil, terodiline, or verapamil. In an embodiment, the peripheral vasodilator is dipyridamole, nylidrin hydrochloride, or isoxsuprine hydrochloride. In an embodiment, the cinchona alkaloids is quinidine or quinine. In an embodiment, the amide is procainamide, flecainide, lidocaine, bupivicaine, ropivicaine, or dispyramide. In an embodiment, the diidobenzyloxyethylamine is amiodarone. In an embodiment, the hydantoin is phenytoin. In an embodiment, the diuretic is a thiazide diuretic, an osmotic diuretic, a carbonic anhydrase inhibitor, a benzothiadiazide diuretic, a loop diuretic, or a potassium sparing diuretic. In an embodiment, the thiazide diuretic is bendrofiuazide, bendroflumethiazide, benzthiazide, benzylhydrochlorothiazide, chlorothiazide, chlorthalidone, cyclopenthiazide, diucardin, diuril, enduron, esidrix, exna, HCTZ, hydrochlorothiazide, hydrodiuril, hydroflumethiazide, bendroflumethiazide, hydromox, hygroton, indapamide, lozol, methylchlorthiazide, metolazone, mykrox, naqua, naturetin, oretic, penflutizide, polythiazide, quinethazone, renese, trichlormethiazide, xipamide, or zaroxolyn. In an embodiment, the osmotic diuretic is mannitol, urea, glycerin, or isorbide. In an embodiment, the carbonic anhydrase inhibitor is acetazolamide. In an embodiment, the benzothiadiazide diuretic is or chlorothiazide. In an embodiment, the loop diuretic is azosemide, furosemide, bumetanide, ethacrynic acid, piretanide, or torasemide. In an embodiment, the potassium sparing diuretic is triamterene, amiloride, spironolactone, or eplerenone. In an embodiment, the vasodilator is bencyclane, cinnarizine, citicoline, cyclandate, cyclonicate, diazoxide, ebumamonine, flunarizine, hydralazine, ibudilast, ifenprodil, lomerizine, minoxidil, naphlole, nikamate, nimodipine, nitroprusside, nofedoline, nosergoline, papaverine, pentifylline, pentoxifylline, phenoxezyl, prostacyclin derivatives, vichizyl, vincamin, or vinpocetine. In an embodiment, the peripheral sympatholytic is acebutol, atenolol, betaxolol, bisoprolol, carteolol, carvediolol, doxazosin, guanadrel, guanethidine, labetalol, metoprolol, nadolol, penbutolol, pindolol, prazosin, propanolol, reserpine, terazosin, or timolol. In an embodiment, the central α2-sympathomimetic is clonidine, guanabenz, guanfacine, or methyldopa. In an embodiment, the angiotensin-converting enzyme inhibitor is alacepril, benazepril, captopril, ceranopril, cilazopril, delapril, enalapril, fosinopril, imidapril, lisinopril, mobertpril, moexipril, pentopril, perindopril, quinapril, ramipril, randolapril, spirapril, temocapril, trandolapril, or zofenopril. In an embodiment, the angiotensin II- receptor antagonist is candesartan, eprosartan, ibersartan, losartan, telmisartan, or valsartan. In an embodiment, the HMG CoA reductase inhibitor is livostatin, simvastatin, pravastatin, fluvastatin, atorvastatin, or cerivastatin. In an embodiment, the fibrate is nicotinic acid, bezafibrate, clinofibrate, clofibrate, colestimide, fenofϊbrate, gemfibrizil, probucol, or simfibrate. In an embodiment, the antiarrhythmic is tocainide, encainide, phenytoin, flecainide, tocainide, propafenone, bretylium, sotalol, ibutilide, dofetilide, bepridil, moricizine, propanolol, esmolol, artilide, clofϊlium, azimilide, dronedarone, ersentilide, ibutilide, tedisamil, trecetilide, verapamil, diltaizem, digitalis, adenosine, nickel chloride, procainaltide, lignocaine, or magnesium ions. In an embodiment, the cardiac glycoside is allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, a xanthine formulation, a catecholamine formulation, denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, or colforsin daropate. In an embodiment, the xanthine formulation is aminophylline, choline theophylline, diprophylline, or proxphylline. In an embodiment, the catecholamine formulation is dopamine, dobutamine, or docarpamine. In an embodiment, the heparin agent is heparin sodium, heparin potassium, a low-molecular weight heparin fragment, or a low-molecular weight heparinoid. In an embodiment, the low-molecular weight heparin fragment is enoxaparin, dalteparin, tinzaparin, or ardeparin. In an embodiment, the low-molecular weight heparinoid is danaparoid. In an embodiment, the coumarin derivative is warfarin or dicumarol. In an embodiment, the histone deacetylase inhibitor is a class I and/or a class II histone deacetylase inhibitor. In an embodiment, the inhibitor of the class I and'or class II histone deacetylase is a small molecular weight carboxylate. In an embodiment, the inhibitor of the class I and/or class II histone deacetylase is a hydroxamic acid. In an embodiment, the inhibitor of the class I and/or class II histone deacetylase is a benzamide. In an embodiment, the inhibitor of the class 1 and/or class 11 histone deacetylase is an epoxyketone. In an embodiment, the inhibitor of the class I and/or class II histone deacetylase is a cyclic peptide. In an embodiment, the inhibitor of the class I and/or class II histone deacetylase is a hybrid molecule. In an embodiment, the inhibitor of the class I and'Or class II histone deacetylase is Suberoylanilide Hydroxamic Acid (SAHA (e.g., MK0683, vorinostat) and other hydroxamic acids), Suberoyl bis-hydroxamic Acid, BML-210, Depudecin (e.g., (-)- Depudecin), HC Toxin, Nullscript (4-(l,3-Dioxo-lH,3H-benzo[de]isoqumolm-2-yl)-N- hydroxybutanamide), Phenylbutyrate (e.g., sodium phenylburyrate) and Valproic Acid (and other short chain fatty acids), Scriptaid, Splitomicin, Suramin Sodium, Trichostatin A (TSA), APHA Compound 8, Apicidin, Sodium Butyrate, pivaloyloxymethyl butyrate (Pivanex, AN- 9), Trapoxin B, Chlamydocin, Depsipeptide (also known as FR901228 or FK228), benzamides (e.g., CI-994 (i.e., N-acetyl dinaline) and MS-27-275), MGCDOl 03, NVP-LAQ-824, CBHA (m-carboxycinnaminic acid bishydroxamic acid), JNJ16241199, Tubacin, A-161906, proxamide, oxamflatin, 3-Cl-UCHA (i.e., 6-(3- chlorophenylureido)caproic hydroxamic acid), AOE (2-amino-8-oxo-9,10-epoxydecanoic acid), CHAP31 or CHAP 50. In an embodiment, the SIRT inhibitor is a 2- anilinobenzamide or 2-chloro-5,6,7,8,9,10-hexahydrocyclohepta[b]indole-6, 2-chloro- 5,6,7,8,9,10-hexahydrocyclohepta[b]indole-6-carboxylate, or 2-chloro-5, 6,7,8, 9,10- hexahydrocyclohepta[b]indole-6-carboxamide, or 6-Chloro-2,3 ,4,9-tetrahydro- 1 H- carbazole-1-carboxylic acid amide. In an embodiment, the 2-anilinobenzamide is nicotinamide, carba-NAD+, EX-527, sirtinol, para-sirtinol, JFD00244, splitomicin, HR73, or cambinol. In an embodiment, the xanthine oxidase inhibitor is allopurinol. In an embodiment, the alkylating agent is altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, or thiotepa. In an embodiment, the antitumor antibiotic is bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitoxantrone, or valrubicin. In an embodiment, the antimetabolite is capecitabine, cladribine, cytarabine, fludarabine, floxuridine, 5-fluorouracil, gemicabine, hydroxyurea, 6-mercaptopurine, methotrexate, pentostatin, or 6-thioguanine. In an embodiment, the mitotic inhibitor is docetaxel, paclitaxel, vinblastine, vincristine, or vinorelbine. In an embodiment, the topoisomerase inhibitor is etoposide, irinotecan, teniposide, or topotecan. In an embodiment, the enzyme is asparaginase or pagasparaginase. In an embodiment, the protein tyrosine kinase inhibitor is imatinib mesylate. In an embodiment, the hormonal agent is an adrenocorticoid, an estrogen, a progestin, an antiestrogen, an androgen, or an antiandrogen. In an embodiment, the adrenocorticoid is dexamethasone, methylprednisolone, or prednisone. In an embodiment, the estrogen is diethylstilbestrol or estradiol. In an embodiment, the progestin is medroxyprogesterone or megesterol acetate. In an embodiment, the antiestrogen is fulvestrant, tamoxifen, or toremifene. In an embodiment, the androgen is testosterone, methyltestosterone, or fluoxymesterone. In an embodiment, the antiandrogen is bicalutamide, flutamide, or nilutamide. In an embodiment, the aromatase inhibitor is aminoglutethimide, anastrozole, exemestane, or letrozole. In an embodiment, the LHRH analogue is leuprolide or goserelin. In an embodiment, the biological agent is interferon-α 2a and 2b, interleukin-2, interleukin-11, the growth factors filgrastim, pegfϊlgrastim, sargramostim, epoetin alfa, darbepoetin alfa, or a monoclonal antibody. In an embodiment, the benzodiazepine is diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, and xanax. In an embodiment, the opioid-receptor antagonist is tramadol or naltrexone. In an embodiment, the antidepressant is a selective serotonin reuptake inhibitor, a serotonin and norepinephrine reuptake inhibitor, a tetracyclic antidepressant, a norepinephrine and dopamine reuptake inhibitor, a combined reuptake inhibitor and receptor blocker, a tricyclic antidepressant, or a monoamine oxidase inhibitor. In an embodiment, the selective serotonin reuptake inhibitor is citalopram, escitalopram, fluoxetine, paroxetine, or sertraline. In an embodiment, the serotonin and norepinephrine reuptake inhibitor is duloxetine or venlafaxine. In an embodiment, the tetracyclic antidepressant is mirtazapine. In an embodiment, the combined reuptake inhibitor and receptor blocker is trazodone, nefazodone, or maprotiline. In an embodiment, the tricyclic antidepressant is amitriptyline, amoxapine, desipramine, doxepin, imipramine, nortriptyline, protriptyline, or trimipramine. In an embodiment, the monoamine oxidase inhibitor is phenelzine, ranylcypromine, isocarboxazid, or selegiline. In an embodiment, the analgesic is paracetamol, a non-steroidal anti-inflammatory drug, a cyclooxygenase-2 inhibitor, an opiate, a corticosteroid, or a topical analgesic. In an embodiment, the paracetamol is acetaminophen or indomethacin. In an embodiment, the non-steroidal anti-inflammatory drugs comprise, aspirin, ibuprofen, salsalate, magnesium salicylate, or a COX-2 selective inhibitor. In an embodiment, the COX -2 selective inhibitor is celecoxib, refecoxib, or valdecoxib. In an embodiment, the opiate is codeine, oxycodone, hydrocodone, diamorphine, pethidine, tramadol, or buprenorphine. In an embodiment, the corticosteroid is prednisone, prednisolone, cortisone, or hydrocortisone. In an embodiment, the topical analgesic is capsaicin. In an embodiment, the amphetamine is dextroamphetamine, benzedrine, ritalin, phenmetrazine, phendimetrazine, methamphetamine, diethylpropion, phentermine, sibrutamine, or orlistat.
In another aspect, the invention features a method of treating a disorder in a subject. The method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent. In an embodiment, the disorder is obesity. In a further embodiment, the disorder is a combination of disorders. In an embodiment, the combination of disorders is obesity and syndrome X. In another embodiment, syndrome X is diabetes. In an embodiment, the additional therapeutic agent is an anti-diabetic agent. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-diabetic agent. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered in a single dosage form. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are provided in amounts so as to provide a synergistic effect. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered one time daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered twice daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered three times daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered within 1 hour before or two hours after a meal. In an embodiment, the ghrelin antagonist, for example a compound of formula 1, II, III, IV, V, or VI and the anti-diabetic agent are administered without regard to food.
In another aspect, the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- diabetic agent.
In another aspect, the invention features a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-diabetic agent. In an embodiment, the solid dosage form includes a tablet or capsule. In an embodiment, the solid dosage form includes an enteric coating. In an embodiment, the anti-diabetic agent is insulin, tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, glimepiride, repaglinide, nateglinide, acarbose, miglitol, metformin, pioglitazone, or rosiglitazone. In an embodiment, the insulin is short acting insulin, regular insulin, semilente insulin, intermediate acting insulin, lente insulin, or long acting insulin. In another aspect, the invention features a method of treating a disorder in a subject. The method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent. In an embodiment, the disorder is obesity. In a further embodiment, the disorder is a combination of disorders. In an embodiment, the combination of disorders is obesity and cardiovascular disease. In an embodiment, wherein the additional therapeutic agent is a cardiovascular disease treating agent. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with a cardiovascular disease treating agent. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered in a single dosage form. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are provided in amounts so as to provide a synergistic effect. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered one time daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered twice daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered three times daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered within 1 hour before or two hours after a meal. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered without regard to food. In aother aspect, the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent. In an embodiment, the pharmaceutical composition is in a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent. In an embodiment, the solid dosage form includes a tablet or capsule. In an embodiment, the tablet or capsule includes an enteric coating. In an embodiment, the cardiovascular treating agent is, allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, aminophylline, choline theophylline, diprophylline, proxphylline, dopamine, dobutamine, docarpamine, denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, colforsin daropate, amyl nitrite, nitroglycerin, propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, carteolol, oxprenolol, penbutolol, levatol, medroxalol, bucindolol, levobunolol, metipranolol, bisoprolol, nebivolol, betaxolol, celiprolol, sotalol, propafenone, verapamil, diltiazem, nicardipine, nifedipine, isradipine, amlodipine, felodipine, nisoldipine, nimodipine, bepridil, dipyridamole, nylidrin hydrochloride, isoxsuprine hydrochloride, quinidine, procainamide, flecainide, lidocaine, bupivicaine, ropivicaine, dispyramide, lidocaine, mexiletine, bretylium, amiodarone, phenytoin, tocainide, encainide, phenytoin, flecainide, tocainide, propafenone, bretylium, sotalol, ibutilide, dofetilide, bepridil, moricizine, propanolol, esmolol, artilide, clofilium, azimilide, dronedarone, ersentilide, ibutilide, tedisamil, trecetilide, verapamil, diltaizem, digitalis, adenosine, nickel chloride, procainaltide, lignocaine, magnesium ions, bendrofluazide, bendroflumethiazide, benzthiazide, benzylhydrochloro thiazide, chlorothiazide, chlorthalidone, cyclopenthiazide, diucardin, diuril, enduron, esidrix, exna, HCTZ, hydrochlorothiazide, hydrodiuril, hydroflumethiazide, bendroflumethiazide, hydromox, hygroton, indapamide, lozol, methylchlorthiazide, metolazone, mykrox, naqua, naturetin, oretic, penflutizide, polythiazide, quinethazone, renese, trichlormethiazide, xipamide, zaroxolyn, mannitol, urea, glycerin, isorbide, acetazolamide, chlorothiazide, azosemide, furosemide, bumetanide, ethacrynic acid, piretanide, torasemide, triamterene, amiloride, spironolactone, eplerenone, bencyclane, cinnarizine, citicoline, cyclandate, cyclonicate, diazoxide, ebumamonine, flunarizine, hydralazine, ibudilast, ifenprodil, lomerizine, minoxidil, naphlole, nikamate, nimodipine, nitroprusside, nofedoline, nosergoline, papaverine, pentifylline, pentoxifylline, phenoxezyl, prostacyclin derivatives, vichizyl, vincamin, vinpocetine, acebutol, atenolol, betaxolol, bisoprolol, carteolol, carvediolol, doxazosin, guanadrel, guanethidine, labetalol, metoprolol, nadolol, penbutolol, pindolol, prazosin, propanolol, reserpine, terazosin, timolol, clonidine, guanabenz, guanfacine, methyldopa, amlodipine, arandipine, bamidipine, bencyclane, benidipine, bepridil, cilnidipine, cinnarizine, clentiazem, diltiazem, efonidipine, elgodipine, etafenone, flunarizine, filodepine, galopamil, isradipine, lacidipine, lercanidipine, lidoflazine, lomerizine, manidipine, mibefradil, nicardipine, nifedipine, nilvadepine, nimodipine, nisoldipine, nitrendipine, penylamine, perhexiline, phendilin, semotiadil, terodiline, verapamil, candesartan, eprosartan, ibersartan, losartan, telmisartan, valsartan, alacepril, benazepril, captopril, ceranopril, cilazopril, delapril, enalapril, fosinopril, imidapril, lisinopril, mobertpril, moexipril, pentopril, perindopril, quinapril, ramipril, randolapril, spirapril, temocapril, trandolapril, zofenopril, cholestyramine chloride, colestipol hydrochloride, livostatin, simvastatin, pravastatin, fluvastatin, atorvastatin, cerivastatin, nicotinic acid, bezafϊbrate, clinofibrate, clofϊbrate, colestimide, fenofibrate, gemfibrizil, probucol, simfibrate, eicosapentaenoic acid, docosahexaenoic acid, heparin, heparin sodium, heparin potassium, enoxaparin, dalteparin, tinzaparin, ardeparin, danaparoid, lepirudin, warfarin, dicumarol, phenindione, aspirin, indomethacin, ticlopidine, cilostazol, clopidogrel, dipyridamole, tirofϊban, eptifibatide, anagrelide, Fab fragments of human monoclonal antibody to the GPIIb/IIIa receptor, alteplase, reteplase, tenecteplase, streptokinase, anistreplase, or urokinase. In another aspect, the invention features a method of treating a disorder in a subject. The method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent. In an embodiment, the disorder is obesity. In a further embodiment, the disorder is a combination of disorders. In an embodiment, the combination of disorders is obesity and cancer. In an embodiment, wherein the additional therapeutic agent is an anti-cancer agent. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-cancer agent. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anticancer agent are administered in a single dosage form. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are provided in amounts so as to provide a synergistic effect. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anticancer agent are administered one time daily. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered twice daily. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer treating agent are administered three times daily. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered within 1 hour before or two hours after a meal. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered without regard to food. In another aspect, the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anticancer agent. In an embodiment, the pharmaceutical composition is in a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent. In embodiment, the solid dosage form includes a tablet or capsule. In an embodiment, the tablet or capsule includes an enteric coating. In an embodiment, the anticancer agent is, altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, thiotepa, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitoxantrone, valrubicin, capecitabine, cladribine, cytarabine, fludarabine, floxuridine, 5-fluorouracil, gemicabine, hydroxyurea, 6-mercaptopurine, methotrexate, pentostatin, 6-thioguanine, docetaxel, paclitaxel, vinblastine, vincristine, vinorelbine, etoposide, irinotecan, teniposide, topotecan, asparaginase, pagasparaginase, imatinib mesylate, dexamethasone, methylprednisolone, prednisone, diethylstilbestrol, estradiol, medroxyprogesterone, megesterol acetate, fulvestrant, tamoxifen, toremifene, aminoglutethimide, anastrozole, exemestane, letrozole, testosterone, methyltestosterone, fluoxymesterone, bicalutamide, flutamide, nilutamide, leuprolide, goserelin, interferon-α 2a and 2b, interleukin-2, interleukin-11, filgrastim, pegfϊlgrastim, sargramostim, epoetin alfa, darbepoetin alfa, monoclonal antibodies, or oncolytic adenoviruses.
In another aspect, the invention features a method of treating a disorder in a subject. The method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent. In an embodiment, the disorder is obesity. In a further embodiment, the disorder is a combination of disorders. In an embodiment, the combination of disorders is obesity and addiction. In an embodiment, the additional therapeutic agent is an anti-addictive agent. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-addictive agent. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered in a single dosage form. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- addictive agent are provided in amounts so as to provide a synergistic effect. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered one time daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- addictive agent are administered twice daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive treating agent are administered three times daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered within 1 hour before or two hours after a meal. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- addictive agent are administered without regard to food.
In another aspect, the invention features a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent. In one embodiment, the pharmaceutical composition is in a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- addictive agent. In an embodiment, the solid dosage form includes a tablet or capsule. In an embodiment, the tablet or capsule includes an enteric coating. In an embodiment, the anti-addictive agent is, nicotine replacement, diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, xanax, propanolol, clonidine, bupropion hydrochloride, naltrexone, citalopram, fluoxetine, zimelidine, sertraline, paroxetine, disulfiram, naltrexone, nalmefene, acamprosate, buspirone, or ondansetron.
In another aspect, the invention features a method of treating a disorder in a subject. The method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent. In an embodiment, the disorder is obesity. In a further embodiment, the disorder is a combination of disorders. In an embodiment, the combination of disorders is obesity and osteoarthritis. In an embodiment,the additional therapeutic agent is an anti- osteoarthritic agent. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-osteoarthritic agent. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or Vl and an anti-osteoarthritic agent are administered in a single dosage form. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are provided in amounts so as to provide a synergistic effect. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered one time daily. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II,
III, IV, V, or VI and an anti-osteoarthritic agent are administered twice daily. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered three times daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered within 1 hour before or two hours after a meal. In an embodiment,the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered without regard to food.
In another aspect, the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- osteoarthritic agent. In an embodiment, the pharmaceutical composition is in a solid dosage form including a ghrelin antagonist, for example a compound of formula I, II, III,
IV, V, or VI and an anti-osteoarthritic agent. In an embodiment, the solid dosage form includes a tablet or capsule. In an embodiment, the tablet or capsule includes an enteric coating. In an embodiment, the antiosteoarthritic is, acetaminophen, salsalate, magnesium salicylate, aspirin, diclofenac, etodolac, ibuprofen, naproxen, nabumeton, sulindac, tolmetin, celecoxib, refecoxib, valdecoxib, tramadol, codeinek, oxycodone, capsaicin, corticosteroids, hyaluronic acid derivatives, glucosamine, chondroitin, or S- adenosyl-methionine. In another aspect, the invention features a method of treating a disorder in a subject. The method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent. In an embodiment, the additional therapeutic agent is a weight loss promoting agent. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with a weight loss promoting agent. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, 111, IV, V, or VI and a weight loss promoting agent are administered in a single dosage form. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are provided in amounts so as to provide a synergistic effect. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered one time daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered twice daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered three times daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or Vl and a weight loss promoting agent are administered within 1 hour before or two hours after a meal. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered without regard to food. In another aspect, the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent. In an embodiment, the pharmaceutical composition is in a solid dosage form inlcuding a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent. In an embodiment, the solid dosage form includes a tablet or capsule. In an embodiment, the tablet or capsule includes an enteric coating. In an embodiment, the weight loss promoting therapeutic agent is, dexfenfluramine, amphetamine, methamphetamine, phenmetrazine, phendimetrazine, diethylpropion, phentermine, sibutramine, or orlistat.
In another aspect, the invention features a method of treating a disorder in a subject. The method includes administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent. In an embodiment, the disorder is obesity. In a further embodiment, the disorder is a combination of disorders. In an embodiment, the combination of disorders is obesity and gout. In an embodiment, the additional therapeutic agent is an anti-gout agent. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or Vl and an anti-gout agent are administered in a single dosage form. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are provided in amounts so as to provide a synergistic effect. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered one time daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered twice daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered three times daily. In an embodiment, the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered within 1 hour before or two hours after a meal. In an embodiment, the ghrelin antagonist, for example a compound of formula 1, 11, III, IV, V, or VI and an anti-gout agent are administered without regard to food.
In another aspect, the invention features a pharmaceutical composition including a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti- gout agent. In an embodiment, the pharmaceutical composition is in a solid dosage form inlcuding a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent. In an embodiment, the solid dosage form includes a tablet or capsule. In an embodiment, the tablet or capsule includes an enteric coating. In an embodiment, the anti-gout agent is, vorinostat, zolinza, SAHA, depsipeptide, MS-275, MGCD0103, PXDlOl, baceca, savicol, LBH589, PCI-24781, or ITF2357.
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 Cl-C 8 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 aromatic ring system 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 "amino acid" refers to a molecule containing both an amino group and a carboyxl group. Suitable amino acids include, without limitation, both the D- and L- isomers of the 20 naturally occurring amino acids found in peptides (e.g., A, R, N, C, D, Q, E, G, H, I, L, K, M, F, P, S, T, W, Y, V (as known by the one letter abbreviations)) as well as unnaturally occurring amino acids prepared by organic synthesis or other metabolic routes.
The term "amino acid side chain" refers to any one of the twenty groups attached to the α-carbon in naturally occurring amino acids. For example, the amino acid side chain for alanine is methyl, the amino acid side chain for phenylalanine is phenylmethyl, the amino acid side chain for cysteine is thiomethyl, the amino acid side chain for aspartate is carboxymethyl, the amino acid side chain for tyrosine is 4- hydroxyphenylmethyl, etc.
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- 1 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, FOXO-3, or FOXO-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. A "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 FOXO-4. Exemplary IGF-I Receptor signaling pathway effector can include: GADD45, PA26, Selenoprotein P, Whipl, cyclin G2, andNIP3.
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 (ELlSA), radioimmunoassay (RJA) 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 accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims. BRIEF DESCRIPTION OF DRAWINGS
Figures 1 and 2 demonstrate that compounds 57 and 94 block ghrelin-induced food intake in mice. Figures 3a and 3b demonstrate how administration of compounds 57 and 94 resulted in decreased body weight in mice. No apparent effect on food intake was observed.
Figures 4a-4f demonstrate improvement in insulin sensitivity in mice dosed with compounds 57 and 94. Figure 5 demonstrates improved plasma HbAIc levels and improved cholesterol levels at 28 days in mice dosed with compound 57.
Figure 6 demonstrates improved plasma HbAIc levels, improved cholesterol levels and improved triglyceride levels at 28 days in mice dosed with compound 94.
Figure 7 demonstrates improved fasting blood glucose at 14 days in mice dosed with compound 57.
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.
Representative compounds of the invention are depicted below in Table 1. Other exemplary compounds are within the scope set forth in the Summary or are described elsewhere herein.
Table 1: Exemplary GHS-R Modulating Compounds
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
* A refers to a compound having antagonist activity with a Ki < 100 nM in a cell based assay. B refers to a compound having antagonist activity with a Ki between 100 nM and 500 nM in a cell based assay.
C refers to a compound having antagonist activity with a Ki between 500 nM and 1000 nM in a cell based assay.
D refers to a compound having antagonist activity with Ki, < 1000 nM in a cell-based assay.
E refers to other exemplary compounds. Representative compounds that modulate GHS-R include the compounds of formulas
(1), (11), (111), (IV), (V), and (Vl) below, where all variables are as described herein.
Figure imgf000076_0001
formula (1) formula (Tl) formula (TTT)
Figure imgf000076_0002
formula (IV) formula (V) formula (VI).
In some preferred embodiments, Y is a 5 membered heteroaromatic moiety substituted with 1 or 2 substituents as described herein. Exemplary Y moieties are reproduced below.
Figure imgf000076_0003
Figure imgf000076_0004
In each instance, any atom, including the hydrogens depicted on the nitrogen atoms, can be substituted with R10. In some preferred embodiments, the heteroaryl moiety includes 1 or 2 R10 substituents. In some preferred embodiments, R10 is aryl, arylalkyl, or R15. When two R10 substituents are included, in some embodiments, one R10 is R15 and the second R10 is a different substituent, such as alkyl, alkoxy, halo, etc.
In certain instances, R1 is an aryl moietiy such as a phenyl moiety, for example unsubstituted or substituted aryl moiety. In some instances, R1 is a heteroaryl moiety such as an indole moiety. In many instances where R1 is aryl or heteroaryl (or other lipophilic moiety such as alkyl), K is an oxygen or a bond. A and R4 and R5 can be chosen to vary the compound's type of interaction with GHS-
R. For example, in some instances where R4 and R5 are both hydrogen, the compound is an agonist of GHS-R. In other instances where R4 and R5 are both independently alkyl, the compound is an antagonist of GHS-R.
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.
In some embodiments, a Y moiety, or other ring corresponding to a Y moiety, can be synthesized onto an amino acid or amino acid type starting material as depicted in schemes A and B and B' below. Scheme A
Figure imgf000078_0001
In the schemes provided herein, all variables are defined as herein and PG is a nitrogen protecting group. The nitrogen protected amino acid is reacted with a N- hydroxy imidamide (amidoxime) moiety (which is prepared by reacting a cyano containing moiety with hydroxylamine) to produce an oxadiazole containing moiety. The resulting compound can be further manipulated to form a compound of formula (I) by removing the nitrogen protecting group and reacting the resulting moiety with an activated sulfone, such as a sulfonyl chloride as depicted below.
Figure imgf000078_0002
Scheme B below depicts the formation of a triazole containing moiety which can be further reacted in a manner similar to the oxadiazole containing moiety to form a compound of formula (I).
Figure imgf000078_0003
Scheme B
Scheme B' below depicts an alternative method of forming a triazole containing moiety which can be further reacted in a manner similar to the oxadiazole containing moiety to form a compound of formula (I). Ri
Figure imgf000079_0001
Figure imgf000079_0002
The triazole precursor moiety can be prepared in a variety of manners, for example, by reacting a cyano containing moiety with a hydrazine hydrate (to form the intermediate amidrazone) as depicted in scheme B. Alternatively, the triazole precursor moiety can be prepared as shown in scheme B' by reacting a nitrile moiety (e.g., an arylnitrile or benzylnitrile) with a dialkyl dithiophosphate moiety such as diethyl dithiophosphatc to provide an thioimidatc, which is reacted with a acyl hydrazidc moiety to provide the triazole precursor. The acyl hydrazide moiety can be prepared, for example, by reacting a carboxylic acid or derivative thereof with hydrazine.
In other embodiments, a compound of formula (I) can be prepared by first reacting an activated sulfone moiety (e.g., a sulfonyl chloride) with an amino acid moiety or protected amino acid, as depicted in Scheme C below.
Figure imgf000079_0004
Scheme C.
Figure imgf000079_0003
The free carboxyl moiety can then be further manipulated to produce a compound of formula (I). For example, the free carboxyl moiety can be reacted with a compound of formula (X) or (XI) above to form an oxadiazole or triazole containing compound of formula (I) in a manner similar to that described in schemes A, B and B' above.
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 Reagentsor Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof. Additionally, the compounds disclosed herein can be prepared on a solid support or using a solid phase peptide synthesis.
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, Czaraik, A.W., Curr. Opin. Chem. Bio., (1997) 1, 60).
The term "solid phase peptide" refers to an amino acid, which is chemically bonded to a resin (e.g., a solid support). Resins are generally commercially available (e.g., from SigmaAldrich). Some examples of resins include Rink-resins, Tentagel S RAM, MBHA, and BHA-resins. 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. AH 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^1 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, Ca2+ 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 10 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 40C 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 Ki) can be estimated from competition binding experiments using the equation: Kd = ICw-L and K; = IC so / (1 + L I Kd), where L is the concentration of radioactive ligand. Bmaχ 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]-myø-inositol 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 370C 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 370C, 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 trans fected 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 transfection, 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 μl of artificial cerebrospinal fluid for injection. For ICV injection, mice are anaesthetised 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)) χ 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 anaesthesia 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 cDNA probe (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 glyceraldehyde 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 K; (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 SOD 1-G93 A 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 Drυsυphila 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, intraarterial Iy, 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 injectables, 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 an compound effective to prevent a disorder, or a "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. 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 timezones, 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, cardiomyopathy, coronary heart disease, venous thromboembolic disease, endocarditis, congenital heart disease, myocardial ischemia, angina, arrhythmia, diseases of the aorta and its branches, diseases of the peripheral vascular system, orthostatic hypotension, hypertension (i.e., elevated blood pressure), hyperlipidemia, arteriosclerosis, artherosclerosis, myocardial infarction, cardiac fibrillation, congestive heart failure, heart failure, and stroke. 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, elevated cholesterol, or elevated blood pressure. 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. In some embodiments, a compound described herein, for example, a compound of formula I, II, III, IV, V or VI can be used to treat addictive behavior involving, for example, smoking, gambling, and alcoholism.
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-I β, leptin, and gastrin- releasing peptide. Accordingly, the compounds may increase the discharge rate of the gastric vagal afferent.
We have also found that substance P and derivatives thereof can modulate GHS-R activity. In particular, we found that substance P alters feeding activity of mice in the fast refeed assay. Accordingly, substance P and derivatives thereof can be used to modulating an eating or metabolic disorder as well as other disorders described herein.
Our investigation of GHS-R expression in human tissues has demonstrated that GHS-R is 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 neoplaLstic 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 104 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) CrU 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 leukenuVlymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF) and Hodgkin's disease.
Combinations of Compounds described herein with other therapeutic agents In some instances it is desirable to treat a subject with a combination of agents, for example, if a patient is suffering from one conditions having multiple symptoms or is suffering from multiple conditions. For example, obesity is frequently associated with a plurality of inter-related diseases and conditions, such as, diabetes, cardiovascular disease, certain cancers, addictive behavior, and osteoarthritis. Co-administration of two or more therapeutic compounds, therefore could be used to treat obesity and one or more of the above listed associated diseases. In some cases, co-administration of drugs can achieve synergistic results, e.g., greater than additive results. Furthermore, in some cases, a combination (e.g., co-administration) of a plurality of therapeutic agents to treat a plurality of diseases or conditions simultaneously can improve the patient's quality of life by alleviating a plurality of symptoms, for example, symptoms associated with multiple diseases or conditions, and in some embodiments, allowing decreased dosing, which can reduce or eliminate undesirable side effects, due to drug synergism.
As used herein, "co-administration" or "combination therapy" means that two or more agents are administered to a subject at the same time or within a time interval such that there is an overlap of an effect of each agent on the patient. The agents can be administered simultaneously, for example, in a combined unit dose (providing simultaneous delivery of both agents). Alternatively, the agents can be administered at a specified time interval, for example, an interval or minutes, hours, days, or weeks. In general, however, the agents are concurrently bioavailable, e.g., detectable in the subject. In a preferred embodiment, a pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as a compound of Formula I, II, III, IV, V, or VI, may be useful when administered (e.g., coadministered) as part of a combination therapy with an additional therapeutic agent.
Combinations of a ghrelin antagonist and a diabetic therapeutic agent A pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI may be useful when administered (e.g., coadministered) as part of a combination therapy with a second pharmaceutical agent for the simultaneous pharmacotherapy of diabetes and type II diabetes.
Exemplary anti-diabetics that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI include, for example, insulin preparations (e.g., short-acting insulins, such as, lispro insulin, insulin aspart, regular insulin, semilente insulin; intermediate-acting insulins, such as, NPH, lente insulin; and long-acting insulins, such as, PZl, ultrlente insulin, insulin glargine), and oral hypoglycemic agents (e.g., sulfonylureas (e.g., tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, glimepiride), meglitinides (e.g., repaglinide and nateglinide), α-glucosidase inhibitors (acarbose and miglitol), and insulin sensitizers, such as, biguanides (e.g., metformin), and thiazolidinediones (e.g., pioglitazone and rosiglitazone)). In an embodiment, a pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI maybe useful when administered (e.g., coadministered) as part of a combination therapy with metformin.
Combinations of a ghrelin antagonist and an agent used for the treatment of Cardiovascular Disease
Cardiovascular disease (CVD) refers to the group of diseases that affect the heart and/or blood vessels. Exemplary CVDs include, for example, coronary heart disease, venous thromboembolic disease, endocarditis, congenital heart disease, myocardial ischemia, angina, arrhythmia, diseases of the aorta and its branches, diseases of the peripheral vascular system, orthostatic hypotension, hypertension, hyperlipidemia, arteriosclerosis, artherosclerosis, myocardial infarction, cardiac fibrillation, congestive heart failure, heart failure, and stroke. A pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI maybe useful when administered (e.g., co-administered) as part of a combination therapy with one or more pharmaceutical agents directed to the treatment of CVD.
A plurality of pharmaceutical agents, arranged into several classes, are available for the treatment of CVD, for example, cardiac glycosides (cardiotonics), agents for the treatment of myocardial ischemia, antiarrhythmic agents, antihypertensive agents, diuretics, antihyperlipidemic agents, anticoagulant, antiplatelet, thrombolytic agents, and antianemic agents.
Myocardial ischemia, or insufficient blood flow to part of the heart, can cause angina and heart attack. Agents for the treatment of myocardial ischemia include antianginal agents and peripheral vasodilators.
Antianginal agents that may be administered (e.g., coadministered) as apart of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, nitrites, such as amyl nitrite; nitrates, such as nitroglycerin and isosorbide; β-blockers, such as propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, carteolol, oxprenolol, penbutolol, levatol, medroxalol, bucindolol, levobunolol, metipranolol, bisoprolol, nebivolol, betaxolol, celiprolol, sotalol, propafenone; and calcium channel antagonists, such as verapamil, diltiazem, nicardipine, nifedipine, isradipine, amlodipine, felodipine, nisoldipine, nimodipine, and bepridil. Peripheral vasodilators include dipyridamole, nylidrin hydrochloride, and isoxsuprine hydrochloride.
Cardiac dysrhythmia is a group of conditions in which cardiac contractility is faster (tachycardia) or slower (bradycardia) than normal. Fibrillation of the ventricles (ventricular fibrillation), which involves a breakdown in the hearts inherent coordination during contraction, is life threatening.
Antiarrhythmic agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, cinchona alkaloids (e.g., quinidine), amides (e.g., procainamide, flecainide, lidocaine, bupivicaine, ropivicaine, and dispyramide), xylyl derivatives (e.g., lidocaine, and mexiletine), quaternary ammonium salts (e.g., bretylium), diiodobenzyloxyethylamines (e.g., amiodarone), β-b lockers, calcium antagonists, and hydantoins (e.g., phenytoin).
Yet other exemplary antiarrhythmic agents include, for example, tocainide, encainide, phenytoin, flecainide, tocainide, propafenone, bretylium, sotalol, ibutilide, dofctilidc, bcpridil, and moricizinc, propanolol, csmolol, artilidc, clofϊlium, azimilidc, dronedarone, ersentilide, ibutilide, tedisamil, trecetilide, verapamil, diltaizem, digitalis, adenosine, nickel chloride, procainaltide, lignocaine, and magnesium ions.
Persistent hypertension, also known as high blood pressure, is one of the leading predispositions for stroke, heart attack, heart failure, and arterial aneurysm. Antihypertensive agents are classified into seven functionally distinct groups, as follows: First, diuretics; second, vasodilators; third, peripheral symphatholytics; fourth, central α2- sympathomimetics; fifth, calcium channel blockers; sixth, angiotensin-converting enzyme (ACE) inhibitors and angiotensin II -receptor antagonists.
Exemplary antihypertensive agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, thiazide diuretics, such as, bendrofluazide, bendroflumethiazide, benzthiazide, benzylhydrochlorothiazide, chlorothiazide, chlorthalidone, cyclopenthiazide, diucardin, diuril, enduron, esidrix, exna, HCTZ, hydrochlorothiazide, hydrodiuril, hydroflumethiazide, bendroflumethiazide, hydromox, hygroton, indapamide, lozol, methylchlorthiazide, metolazone, mykrox, naqua, naturetin, oretic, penfiutizide, polythiazide, quinethazone, renese, trichlormethiazide, xipamide, zaroxolyn; osmotic diuretics, such as mannitol, urea, glycerin, isorbide; carbonic anhydrase inhibitors, such as acetazolamide; benzothiadiazide diuretics, such as chlorothiazide; loop diuretics, such as azosemide, furosemide, bumetanide, ethacrynic acid, piretanide, torasemide; potassium sparing diuretics, such as, triamterene, amiloride, spironolactone, eplerenone; vasodilators, such as, bencyclane, cinnarizine, citicoline, cyclandate, cyclonicate, diazoxide, ebumamonine, flunarizine, hydralazine, ibudilast, ifenprodil, lomerizine, minoxidil, naphlole, nikamate, nimodipine, nitroprusside, nofedoline, nosergoline, papaverine, pentifylline, pentoxifylline, phenoxezyl, prostacyclin derivatives, vichizyl, vincamin, vinpocetine; peripheral sympatholytics, such as, acebutol, atenolol, betaxolol, bisoprolol, carteolol, carvediolol, doxazosin, guanadrel, guanethidine, labetalol, metoprolol, nadolol, penbutolol, pindolol, prazosin, propanolol, reserpine, terazosin, timolol; central α2-sympathomimetics, such as, clonidine, guanabenz, guanfacine, methyldopa; calcium channel blockers, such as, amlodipine, arandipine, bamidipine, bcncyclanc, bcnidipinc, bcpridil, cilnidipinc, cinnarizinc, clcntiazcm, diltiazcm, efonidipine, elgodipine, etafenone, flunarizine, filodepine, galopamil, isradipine, lacidipine, lercanidipine, lidoflazine, lomerizine, manidipine, mibefradil, nicardipine, nifedipine, nilvadepine, nimodipine, nisoldipine, nitrendipine, penylamine, perhexiline, phendilin, semotiadil, terodiline, verapamil; angiotensin II-receptor antagonists, such as, candesartan, eprosartan, ibersartan, losartan, telmisartan, valsartan; ACE inhibitors, such as, alacepril, benazepril, captopril, ceranopril, cilazopril, delapril, enalapril, fosinopril, imidapril, lisinopril, mobertpril, moexipril, pentopril, perindopril, quinapril, ramipril, randolapril, spirapril, temocapril, trandolapril, and zofenopril.
Hyperlipidemia, which includes hyperlipoproteinemia, hypercholesterolemia, and hypertriglyceridemia, is characterized by elevated serum lipids. Lipids exist in the bloodstream (e.g., cholesterol, cholesterol esters, phospholipids, and triglycerides) as lipoproteins, which are associated with chylomicrons, very low-density lipoproteins (VLDL), intermediate-density lipoproteins (IDL), low-density lipoproteins (LDL), and high-density lipoproteins (HDL). Hyperlipidemia, or more specifically, hypercholesterolemia, is diagnosed in patients with total serum cholesterol greater than 200 mg/dl, or LDL levels greater than 130 mg/dl. Hyperlipidemia causes hardening of arteries, or atherosclerosis; increasing the risk of heart disease, stroke, and other vascular disease.
Antihyperlipidemic agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula 1, 2, 3, or 4, include, for example, nonabsorbable agents (cholestyramine chloride and colestipol hydrochloride) and absorbable agents, including, statins (e.g., livostatin, simvastatin, pravastatin, fluvastatin, atorvastatin (commonly referred to as lipitor), and cerivastatin), fibrates (nicotinic acid, bezafϊbrate, clinofibrate, clofibrate, colestimide, fenofibrate, gemfϊbrizil, probucol, simfibrate), and fatty fish oils containing large amounts of eicosapentaenoic acid and docosahexaenoic acid.
Congestive heart failure results when the heart can no longer pump sufficient supplies of blood to the body's organs. Other conditions that decrease the ejection fraction of the heart include, for example, heart failure, atrial fibrillation, atrial flutter, and paroxysmal atrial tachycardia. Agents used for the treatment of these conditions include the category of agents known as the cardiac glycosides. Cardiac glycosides increase mycocardial contractility and efficiency to improve systemic circulation and renal perfusion. Common cardiac glycosides that may be administered (e.g., coadministered) in combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, xanthine formulations (e.g., aminophylline, choline theophylline, diprophylline, proxphylline), catecholamine formulations (e.g., dopamine, dobutamine, docarpamine), denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, and colforsin daropate.
Patients with an advanced risk of heart disease and stroke may also be treated with combination therapy including a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, and an anticoagulant or antiplatelet agent. Exemplary anticoagulant agents include, for example, heparin (e.g., heparin sodium and heparin potassium) and low-molecular weight heparin fragments (enoxaparin, dalteparin, tinzaparin, ardeparin), low-molecular weight heparinoids (danaparoid), lepirudin, coumarin derivatives (warfarin and dicumarol), and phenindione. Exemplary antiplatelet agents include, for example, cyclooxygenase inhibitors (e.g., aspirin and indomethacin), ticlopidine, cilostazol, clopidogrel, dipyridamole, GPIIb/IIIa receptor antagonists (e.g., tirofiban and eptifϊbatide), anagrelide, and the Fab fragments of human monoclonal antibody to the GPIIb/IIIa receptor (e.g., Abciximab). Thrombolytic agents are used to dissolve clots and reopen arteries or veins as a treatment for heart attack, stroke, deep vein thrombosis, pulmonary embolism, or occlusion of a peripheral artery. Thrombolytic agents that may be administered (e.g., coadministered) as a part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, tissue plasminogen activators (e.g., alteplase, reteplase, and tenecteplase), streptokinase, anistreplase, and urokinase.
Combinations of a ghrelin antagonist and an anti-cancer agent
Cancer is a disease characterized by abnormal cell growth and the formation of tumors or neoplasms, which posses the potential to spread, or metastasize. The risk factors for cancer are multifactorial, and involve both genetic and environmental factors. Obesity is linked to several types of cancer, including, for example, breast cancer, colon cancer, endometrial cancer, kidney cancer, esophageal cancer, and prostate cancer.
A pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may be administered (e.g., coadministered) as part of a combination therapy with an anticancer therapeutic or regimen. Examples of anticancer therapies include palliative, adjuvant, neoadjuvant, and salvage chemotherapy and therapy using biological agents.
Exemplary chemotherapeutic agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, alkylating agents (e.g., altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, and thiotepa), antitumor antibiotics (e.g., bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitoxantrone, and valrubicin), antimetabolites (e.g., capecitabine, cladribine, cytarabine, fludarabine, floxuridine, 5-fluorouracil, gemicabine, hydroxyurea, 6-mercaptopurine, methotrexate, pentostatin, and 6-thioguanine), mitotic inhibitors (e.g., docetaxel, paclitaxel, vinblastine, vincristine, and vinorelbine), topoisomerase inhibitors (e.g., etoposide, irinotecan, teniposide, and topotecan), enzymes (e.g., asparaginase and pagasparaginase), protein tyrosine kinase inhibitors (e.g., imatinib mesylate), and hormonal agents, such as, adrenocorticoids (e.g., dexamethasone, methylprednisolone, and prednisone), estrogens (e.g., diethylstilbestrol and estradiol), progestins (e.g., medroxyprogesterone and megesterol acetate), antiestrogens (e.g., fulvestrant, tamoxifen, and toremifene), aromatase inhibitors (e.g., aminoglutethimide, anastrozole, exemestane, and letrozole), androgens (e.g., testosterone, methyltestosterone, and fluoxymesterone), antiandrogens (bicalutamide, flutamide, and nilutamide), and LHRH analogues (e.g., leuprolide and goserelin).
Exemplary biological agents that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, interferon-α 2a and 2b (Roferon-A and lntron A, respectively), interleukin-2 (Aldesleukin and Proleukin), interleukin-11 (Oprelvekin and Neumega), the growth factor filgrastim (Neupogen), Pegfϊlgrastim (Neulasta), Sargramostim (Leukine), epoetin alfa (Epogen or Procrit), darbepoetin alfa (Aranesp), and monoclonal antibodies, such as, Alemtuzumab, Gemtuzumab ozogamicin, Ibritumomab tuixetan, Rituximab, and Trastuzumab.
A ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, FV, V, or VI, may also be used as part of a combination therapy with herbal anticancer therapies (e.g., essaic tea, hoxsey therapy, iscador, chaparral, and Pau D' Arco), adjunctive herbal therapies, or with oncolytic adenoviruses.
Combination Therapy to Treat Addictive Behavior
Addictive behavior involving, for example, smoking, gambling, and alcoholism, are frequently associated with obesity. A pharmaceutically effective dose of a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may, therefore, be usefully administered (e.g., coadministered) as part of a combination therapy with a second pharmaceutical for the pharmacotherapy of and addictive behavior.
Smoking cessation therapeutics that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, nicotine replacement, benzodiazepines (e.g., diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, and xanax), propanolol, clonidine, bupropion hydrochloride (Zyban and Wellbutrin), and CHANTIX.
Gambling cessation therapeutics that may be administered (e.g., coadministered) as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, naltrexone, selective serotonin-reuptake inhibitor (SSRI) agents (e.g., citalopram, fluoxetine, zimelidine, sertraline, paroxetine), or a combination of Naltrexone and said SSRI agents. Alcoholism therapeutics that may be used as part of a combination therapy with a ghrelin antagonist, for example, compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, disulfiram, naltrexone, nalmefene (REVEX), acamprosate (Campral), buspirone, ondansetron (Zofran), and SSRI agents (e.g., citalopram (Celexa), escitalopram (Lexapro), fluoxetine (Prozac), paroxetine (Paxil), and sertraline (Zoloft).
Combinations of a ghrelin antagonist and an agent used in the treatment of Osteoarthritis
Osteoarthritis, formally known as degenerative joint disease, is a common chronic condition characterized by cartilage degeneration. The major clinical features of osteoarthritis include pain and stiffness, leading to disability.
Obesity is an important risk factor for osteoarthritis in most joints, and in particular, the knee. Furthermore, weight loss is demonstrated to alleviate the symptoms and progression of osteoarthritis. A pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may, therefore, be useful when administered (e.g., coadministered) as part of a combination therapy with an anti-osteoarthritic agent toalleviate the symptoms of osteoarthritis.
Exemplary anti-osteoarthritic agents that may be used as part of a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula 1, 11, 111, IV, V, or Vl, include analgesics, for example, acetaminophen (Tylenol), nonsteroidal anti-inflammatory drugs (NSAIDs), such as, nonacetylated salicylates (e.g., salsalate (Disalcid) and magnesium salicylate (Doan's)), aspirin, diclofenac (Voltaren), etodolac (Lodine), ibuprofen (Motrin and Advil), naproxen (Naprosyn and Aleve), nabumeton (Relafen), sulindac (Clinoril), tolmetin (Tolectin), and COX-2 inhibitors, such as, celecoxib (Celebrex), refecoxib (Vioxx), and valdecoxib (Bextra). Yet further exemplary anti-osteoarthritic agents include, for example, orally dosed tramadol (Ultram) and opiate analgesics (e.g., codeine and oxycodone); topical analgesics (e.g., capsaicin); intra-articular injections of corticosteroids or hyaluronic acid derivatives; adjunctive treatment with glucosamine, chondroitin, or S-adenosyl- methionine.
Combinations of a ghrelin antagonist and an agent for the treatment of gout
A pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may be useful when administered (e.g., coadministered) as part of a combination therapy with an agent used for the treatment of gout, including, for example, allopurinol, vorinostat, Zolinza, SAHA, depsipeptide (Romidepsin), MS-275, MGCDO 103, PXDlOl, Baceca, Savicol, LBH589, PCI-24781 , and ITF2357.
Combinations of a ghrelin antagonist and a weight loss agent
Obesity is associated with numerous other diseases and conditions for which there is no recommended or approved pharmacotherapy, including, for example, sleep apnea and Pickwikian syndrome. Obesity is also associated with abdominal hernia, varicose veins, gall bladder disease, and liver malfunction. In these cases, the most desirable strategy is prophylaxis through weight loss. A pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may be useful when administered (e.g., coadministered) as part of a combination therapy with a weight loss pharmacotherapy. Exemplary weight loss promoting agents include, for example, dexfenfluramine (Redux), amphetamine, methamphetamine, phenmetrazine (Preludin), phendimetrazine (Plegine), diethylpropion (Tenuate), phentermine (Lonamin), sibutramine (Meredia), and orlistat (Xenical). Further still, a pharmaceutically effective dose of a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may be useful when administered (e.g., coadministered) as part of a combination therapy with a second pharmaceutical agent derived from the following classes of pharmaceuticals compounds, including, for example, insulin preparations, oral hypoglycemic agents, peroxisome proliferator-activated receptor gamma (PP ARγ) agonists, nitrates, nitrites, β-blockers, calcium channel antagonists, peripheral vasodilators, cinchona alkaloids, amides, xylyl derivatives, quaternary ammonium salts, diidobenzyloxyethylamines, hydantoins, diuretics, vasodilators, peripheral sympatholytics, central α2-sympathomimetics, angiotensin-converting enzyme (ACE) inhibitors, angiotensin II-receptor antagonists, HMG CoA reductase inhibitors (statins), fibrates, antiarrhythmics, cardiac glycosides, heparin, coumarin derivatives, histone deacetylase (HDAC) inhibitors (e.g., HDAC I/II inhibitors), SIRT inhibitors, alkylating agents, antitumor antibiotics, antimetabolites, mitotic inhibitors, topoisomcrasc inhibitors, enzymes, protein tyrosine kinase inhibitors, hormonal agents, aromatase inhibitors, LHRH analogues, biological agents, herbal remedies, oncolytic adenoviruses, benzodiazepans, opioid-receptor antagonists, antidepressants, analgesics, hyaluronic acid derivatives, and amphetamines.
Insulin preparations that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, short-acting insulins, such as, lispro insulin, insulin aspart, regular insulin, semilente insulin; intermediate-acting insulins, such as, NPH, lente insulin; and long-acting insulins, such as, PZI, ultrlente insulin, and insulin glargine.
Oral hypoglycemic agents that may be administered in combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, FV, V, or VI, include, for example, sulfonylureas (e.g., tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, glimepiride), meglitinides (e.g., repaglinide, nateglinide, and mitiglinide), α-glucosidase inhibitors (acarbose and miglitol), and insulin sensitizers, such as, biguanides (e.g., metformin), and thiazolidinediones (e.g., pioglitazone and rosiglitazone).
Peroxisome proliferator-activated receptor gamma (PPARγ) agonists, that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, thiazolidinediones (e.g., pioglitazone and rosiglitazone).
Nitrates that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, nitroglycerin (e.g., glyceryl trinitrate, Nitro- Bid, Nitro-Stat, Nitrol, Nitro-Dur), isorbide dinitrate (e.g., Isordil, Sorbitrate, Dilatrate- SR), isorbide-5-mononitrate (e.g., Imdur, ISMO), and erythrityl tetranitrate (e.g., Cardilate).
Nitrites that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, amyl nitrate. β-blockcrs that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, carteolol, oxprenolol, penbutolol, levatol, medroxalol, bucindolol, levobunolol, metipranolol, bisoprolol, nebivolol, betaxolol, celiprolol, sotalol, and propafenone.
Calcium channel antagonists that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, amlodipine, arandipine, bamidipine, bencyclane, benidipine, bepridil, cilnidipine, cinnarizine, clentiazem, diltiazem, efonidipine, elgodipine, etafenone, flunarizine, filodepine, galopamil, isradipine, lacidipine, lercanidipine, lidoflazine, lomerizine, manidipine, mibefradil, nicardipine, nifedipine, nilvadepine, nimodipine, nisoldipine, nitrendipine, penylamine, perhexiline, phendilin, semotiadil, terodiline, and verapamil. Peripheral vasodilators that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, dipyridamole, nylidrin hydrochloride, and isoxsuprine hydrochloride.
Cinchona alkaloids that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, quinidine and quinine.
Amides that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, procainamide, flecainide, lidocaine, bupivicaine, ropivicaine, and dispyramide. Xylyl derivatives that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, lidocaine and mexiletine.
Diidobenzyloxyethylamines that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, amiodarone.
Hydantoins that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, phenytoin.
Diuretics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, thiazide diuretics, such as, bendrofluazide, bendroflumethiazide, benzthiazide, benzylhydrochlorothiazide, chlorothiazide, chlorthalidone, cyclopenthiazide, diucardin, diuril, enduron, esidrix, exna, HCTZ, hydrochlorothiazide, hydrodiuril, hydroflumethiazide, bendroflumethiazide, hydromox, hygroton, indapamide, lozol, methylchlorthiazide, metolazone, mykrox, naqua, naturetin, oretic, penfiutizide, polythiazide, quinethazone, renese, trichlormethiazide, xipamide, zaroxolyn; osmotic diuretics, such as mannitol, urea, glycerin, isorbide; carbonic anhydrase inhibitors, such as acetazolamide; benzothiadiazide diuretics, such as chlorothiazide; loop diuretics, such as azosemide, furosemide, bumetanide, ethacrynic acid, piretanide, torasemide; potassium sparing diuretics, such as, triamterene, amiloride, spironolactone, and eplerenone. Vasodilators that may be administered in a combination therapy with a ghrelin antagonist, for example, a compounds described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, bencyclane, cinnarizine, citicoline, cyclandate, cyclonicate, diazoxide, ebumamonine, flunarizine, hydralazine, ibudilast, ifenprodil, lomerizine, minoxidil, naphlole, nikamate, nimodipine, nitroprusside, nofedoline, nosergoline, papaverine, pentifylline, pentoxifylline, phenoxezyl, prostacyclin derivatives, vichizyl, vincamin, and vinpocetine.
Peripheral sympatholytics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, acebutol, atenolol, betaxolol, bisoprolol, carteolol, carvediolol, doxazosin, guanadrel, guanethidine, labetalol, metoprolol, nadolol, penbutolol, pindolol, prazosin, propanolol, reserpine, terazosin, and timolol.
Central ^-sympathomimetics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, clom'dine, guanabenz, guanfacine, and methyldopa.
Angiotensin-converting enzyme (ACE) inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, alacepril, benazepril, captopril, ceranopril, cilazopril, delapril, enalapril, fosinopril, imidapril, lisinopril, mobertpril, moexipril, pentopril, perindopril, quinapril, ramipril, randolapril, spirapril, temocapril, trandolapril, and zofenopril.
Angiotensin II-receptor antagonists that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, candesartan, eprosartan, ibersartan, losartan, telmisartan, and valsartan.
HMG CoA reductase inhibitors (statins) that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, livostatin, simvastatin, pravastatin, fluvastatin, atorvastatin (commonly referred to as lipitor), and cerivastatin.
Fibrates that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, nicotinic acid, bezafibrate, clinofibrate, clofibrate, colestimide, fenofibrate, gemfibrizil, probucol, and simfibrate.
Antiarrhythmic agents that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, tocainide, encainide, phenytoin, flecainide, tocainide, propafenone, bretylium, sotalol, ibutilide, dofetilide, bepridil, and moricizine, propanolol, esmolol, artilide, clofilium, azimilide, dronedarone, ersentilide, ibutilide, tedisamil, trecetilide, verapamil, diltaizem, digitalis, adenosine, nickel chloride, procainaltide, lignocaine, and magnesium ions.
Cardiac glycosides that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, xanthine formulations (e.g., aminophylline, choline theophylline, diprophylline, proxphylline), catecholamine formulations (e.g., dopamine, dobutamine, docarpamine), denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, and colforsin daropate.
Heparin agents that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, heparin sodium and heparin potassium, low- molecular weight heparin fragments (enoxaparin, dalteparin, tinzaparin, ardeparin), and low-molecular weight heparinoids (danaparoid).
Coumarin derivatives that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, warfarin and dicumarol. Histone deacetylase (HDAC) inhibitors may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as a compound of Formula I, II, III, IV, V, or VI. A number of structural classes of negative regulators of HDACs (e.g., HDAC inhibitors such as HDAC I/II inhibitors) have been developed, for example, small molecular weight carboxylates (e.g., less than about 250 amu), hydroxamic acids, benzamides, epoxyketones, cyclic peptides, and hybrid molecules. (See for example, Drummond DC, Noble CO, Kirpotin DB, Guo Z, Scott GK, et al. (2005) Clinical development of histone deacetylase inhibitors as anticancer agents. Annu Rev Pharmacol Toxicol 45: 495-528, (including specific examples therein) which is hereby incorporated by reference in its entirety). Non-limiting examples of negative regulators of type I/II HDACs include: Suberoylanilide Hydroxamic Acid (SAHA (e.g., MK0683, vorinostat) and other hydroxamic acids), Suberoyl bis- hydroxamic Acid, BML-210, Depudecin (e.g., (-)-Depudecin), HC Toxin, Nullscript (4- ( 1 ,3 -Dioxo- 1 H,3H-benzo[de]isoquinolin-2-yl)-N-hydroxybutanamide), Phenylbutyrate (e.g., sodium phenylbutyrate) and Valproic Acid (and other short chain fatty acids), Scriptaid, Suramin Sodium, Trichostatin A (TSA), APHA Compound 8, Apicidin, Sodium Butyrate, pivaloyloxymethyl butyrate (Pivanex, AN-9), Trapoxin B,
Chlamydocin, Depsipeptide (also known as FR901228 or FK228), benzamides (e.g., CI- 994 (i.e., N-acetyl dinaline) and MS-27-275), MGCD0103, NVP-LAQ-824, CBHA (m- carboxycinnaminic acid bishydroxamic acid), JNJ16241199, Tubacin, A-161906, proxamide, oxamflatin, 3-Cl-UCHA (i.e., 6-(3-chlorophenylureido)caproic hydroxamic acid), AOE (2-amino-8-oxo-9,10-epoxydecanoic acid), CHAP31 and CHAP 50. Other inhibitors include, for example, dominant negative forms of the HDACs (e.g., catalytically inactive forms) siRNA inhibitors of the HDACs, and antibodies that specifically bind to the HDACs. Inhibitors are available, e.g., from BIOMOL International, Fukasawa, Merck Biosciences, Novartis, Gloucester Pharmaceuticals, Aton Pharma, Titan Pharmaceuticals, Schering AG, Pharmion, MethylGene, and Sigma Aldrich.
Sirtuin SIRT inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, 2-anilinobenzamides (nicotinamide, carba-NAD+, EX-527, sirtinol, para-sirtinol, JFD00244, splitomicin, HR73, cambinol) and indoles (Napper A., et al. (2005). J. Med. Chem: 48; 8045-8054 and US 2006/0074124, the contents of each of which are incorporated herein by reference), including, for 2-chloro-5,6,7,8,9,10-hexahydrocyclohepta[b]indole-6, 2-chloro- 5,6,7, 8,9,10-hexahydrocyclohepta[b]indole-6-carboxylate, or 2-chloro-5,6,7,8,9,10- hexahydrocyclohepta[b]indole-6-carboxamide, 6-Chloro-2,3,4,9-tetrahydro-lH- carbazole- 1 -carboxylic acid amide .
Xanthine oxidase inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, Allopurinol.
Alkylating agents that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula 1, H, 111, IV, V, or VI, include, for example, altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, and thiotepa. Antitumor antibiotics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitoxantrone, and valrubicin. Antimetabolites that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, capecitabine, cladribine, cytarabine, fludarabine, floxuridine, 5-fluorouracil, gemicabine, hydroxyurea, 6-mercaptopurine, methotrexate, pentostatin, and 6-thioguanine. Mitotic inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, docetaxel, paclitaxel, vinblastine, vincristine, and vinorelbine.
Topoisomerase inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, etoposide, irinotecan, teniposide, and topotecan.
Enzymes that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, asparaginase and pagasparaginase.
Protein tyrosine kinase inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, FV, V, or VI, include, for example, imatinib mesylate.
Hormonal agents that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula 1, II, III, IV, V, or Vl, include, for example, adrenocorticoids (e.g., dexamethasone, methylprednisolone, and prednisone), estrogens (e.g., diethylstilbestrol and estradiol), progestins (e.g., medroxyprogesterone and megesterol acetate), antiestrogens (e.g., fulvestrant, tamoxifen, and toremifene), androgens (e.g., testosterone, methyltestosterone, and fiuoxymesterone) and antiandrogens (e.g., bicalutamide, flutamide, and nilutamide).
Aromatase inhibitors that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, aminoglutethimide, anastrozole, exemestane, and letrozole.
LHRH analogues that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, leuprolide and goserelin.
Biological agents that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, TV, V, or VI, include, for example, interferon-α 2a and 2b (Roferon-A and Intron A, respectively), interleukin-2 (Aldesleukin and Proleukin), interleukin-11 (Oprelvekin and Neumega), the growth factor filgrastim (Neupogen), Pegfilgrastim (Neulasta), Sargramostim (Leukine), epoetin alfa (Epogen or Procrit), darbepoetin alfa (Aranesp), and monoclonal antibodies, such as, Alemtuzumab, Gemtuzumab ozogamicin, Ibritumomab tuixetan, Rituximab, and Trastuzumab. A ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, may also be administered in a combination therapy with an herbal therapy (e.g., essaic tea, hoxsey therapy, iscador, chaparral, and Pau D' Arco) or with an oncolytic adenovirus. Benzodiazepines that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, and xanax.
Opioid-receptor antagonists that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, Ul, IV, V, or VI, include, for example, tramadol and naltrexone.
Antidepressants that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, selective serotonin reuptake inhibitors (SSRIs) (e.g., citalopram (Celexa), escitalopram (Lexapro), fluoxetine (Prozac), paroxetine (Paxil), sertraline (Zoloft), serotonin and norepinephrine reuptake inhibitors (SNRIs) (e.g., duloxetine (Cymbalta) and venlafaxine (Effexor), norepinephrine and dopamine reuptake inhibitors (NDRIs) (e.g., Wellbutrin and Zyban), tetracyclic antidepressants (e.g., mirtazapine (Remeron), combined reuptake inhibitors and receptor blockers (trazodone, nefazodone, and maprotiline), tricyclic antidepressants (e.g., amitriptyline, amoxapine, desipramine (Norpramin), doxepin (Sinequan), imipramine (Tofranil), nortriptyline (Pamelor), protriptyline (Vivactil), trimipramine (Surmontil), and monoamine oxidase inhibitors (MAOIs) (e.g., phenelzine (Nardil), tranylcypromine (Parnate), isocarboxazid (Marplan), and selegiline (Emsam).
Analgesics that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, paracetamol (acetaminophen and indomethacin), non-steroidal anti-inflammatory drugs (NSAIDs) (aspirin, ibuprofen, salsalate, and magnesium salicylate), and cyclooxygenase-2 (COX-2) inhibitors (e.g., celecoxib (Celebrex), refecoxib (Vioxx), and valdecoxib (Bextra)), opiates (e.g., codeine, oxycodone, hydrocodone, diamorphine, pethidine, tramadol, and buprenoφhine), corticosteroids (e.g., prednisone, prednisolone, cortisone, and hydrocortisone), and topical analgesics, such as, capsaicin.
Amphetamines and sympathomimetic amines that may be administered in a combination therapy with a ghrelin antagonist, for example, a compound described herein, such as, a compound of Formula I, II, III, IV, V, or VI, include, for example, dextroamphetamine, benzedrine, ritalin, phenmetrazine (Preludin), phendimetrazine (Plegine), methamphetamine, diethylpropion (Tenuate), phentermine (Lonamin), sibrutamine (Meredia), and orlistat (Xenical).
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 treat one or more of the following disorders: cachexia, wasting, stimulating growth hormone release in elderly humans, patients with cancer, heart failure, or AIDS; treating growth hormone deficient adults; prevention of cataboHc 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 ostcochondrody-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.
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 an 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: Preparation of N,N-3-Diethylaminopropanesulfonyl chloride hyrochloride (3)
Figure imgf000127_0001
1. 2. 3.
Diethylamine (10ml,96.78mmol) was added slowly to the 1 ,3 Propane sultone (3.94g, 32.26mmol) whilst stirring at O0C. This was allowed to stir overnight whilst warming to ambient temperatures. The reaction mixture was then diluted with MeOH (100ml) followed by the addition of Ambersep900 resin (3Og, 75mmol) and shaken for 2hours. The resin was washed, on a glass sinter with 30rnl aliquots of MeOH (washings discarded). The resin was then treated with 2M HCl(aq) (50ml) for 30mins, and then filtered. The resin was washed with 2M HCl(aq) (2 x 50ml). The combined acidic solution was evaporated in vacuo to give the crude product N,N-3-diethylaminopropane sulfonic acid hydrochloride (2) as a white solid.
Example 2: Synthesis of (R)-3-(benzyloxy)-2-(3- (diethylamino*)propylsiilfonamido)propanoic acid Thionyl chloride (50ml, 685mmol) was added to N,N-3-diethylammopropane sulfonic acid hydrochloride (2) and heated to reflux whilst stirring for 8hours. The solution was allowed to cool to ambient temperatures and then evaporated in vacuo to give N,N-3-diethylaminopropanesulfonyl chloride hyrochloride (3) as a grey/white, very hydroscopic solid (7.9g, 92%). The product was stored under a nitrogen atmosphere.
Figure imgf000127_0002
Synthesis of 4:
O-Bn-D-serine (3g, 15.36 mmol) was suspended in DMF (60 mL). BSTFA (bis(trimethylsilyl)trifluoroacetamide) (0.75 mL, 15.36 mmol) was added to the suspension, which was stirred at room temperature for 2hours. All the starting material was dissolved.
3-Chloropropane sulfonyl chloride (2.25 mL, 18.43 mmol) was then added and the mixture was stirred at room temperature over night. The DMF was removed by evaporation in vacuo and ethyl acetate was added. The desired 4 was extracted using aqueous NaOH (IM) (7OmL). The aqueous phase was then acidified with aqueous HCl (5M) to pH 1. The desired compound was then extracted using ethyl acetate, dried over Na2SO4 and the organic solvent was evaporated in vacuo to give m=3.8g of 4 as a dark brown oil. 1H NMR (CDCl3): 2.27 (m, 2H); 2.90-3.23 (m, 2H); 3.58 (t, 2H); 3.77 (m, IH); 3.90 (m, IH); 3.31 (bs, IH); 4.54 (s, 2H); 5.50 (bs, IH); 7.26-7.36 (m, 5H).
Synthesis of 5:
4 (1.2g, 3.58 mmol), was dissolved in THF (3mL), 3 mL of diethylamine was then added to the solution which was heated at 9O0C in a sealed tube over night. The THF was removed in vacuo and ethyl acetate was added. The desired compound was then extracted using H2O (2X30 mL). The water was then removed in vacuo in the presence of toluene to give m= 1. Ig (82% yield) of a light brown compound.
The oil was dissolved in 100ml of MeOH, and 5g of resin Ambersep 900 was added. The mixture was shaken for Ih and the solvent was removed under reduced pressure. The resin was then washed with MeOH (20ml) and suspended in HCl (2N) for Ih. The resin was filtered and the filtrate evaporated to afford 5 as a yellow oil, 1H NMR showed less than 10% diethylamine. 1H NMR (D2O): 1.04 (t, 6H); 1.84 (m, 2H); 2.47- 2.60 (m, 6H); 2.89 (t, 2H); 3.62 (m, IH); 3.75 (m, IH); 3.85 (m, IH); 4.62 (s, 2H); 7.40- 7.47 (m, 5H). Example 3: Synthests of (R)-3-(diethylamino)-N-(l-(3-subsituted-l,2,4-oxadiazol-5- vI)-3-phenyIpropyl)propane-l-suIfonamide and (R)-2-(diethyIamino)-N-(l-(3- substituted-l,2,4-oxadiazol-5-yl)-3-phenylpropyl)ethanesulfonamide R^N 1
Figure imgf000129_0001
Amidoxime preparation (2)
A suspension of the nitrile(l) in EtOH (10ml) and hydroxylamine 50% in water (10ml) was heated to reflux (approx. 1000C) whilst stirring for 6hrs. The reaction mixture was then evaporated to dryness in vacuo. This was then re-evaporated from toluene (10ml) twice to give the crude product.
Oxadiazole formation (4)
i) N-BOC-Homophenylalanine (200mg, 0.72mmol) was dissolved in EtOAc (10ml) and cooled to O0C. To this solution was added CDI (128mg, 0.79mmol) and stirred at O0C for 15mins, and then a further 2hrs whilst warming to ambient temperatures. The amidoxime (2, 0.79mmol) was then added and the reaction mixture was stirred overnight. The reaction mixture was then washed with sat. ΝaHCθ3(aq) (10ml) then brine (10ml) before drying OVCrNa2SO4, filtering and evaporating to dryness in vacuo. ii) The crude intermediate (i) was dissolved in DMF (2ml) and heated in a CEM Discover focussed microwave for lmin at 18O0C. The solvent was then evaporated in vacuo and the crude product was purified by silica gel column chromatography, eluting with a mixture of EtOAc/Heptanes (typically 1/9).
Sulfonamide formation (5)
iii) A solution of TFA (20% in DCM, 5ml) was added to the oxadiazole (4) at O0C and stirred for 30mins, before allowing to warm to ambient temperatures. The reaction was monitored by TLC (EtO Ac/heptanes, 1/1). The reaction was usually complete after lhr stirring at ambient temperatures. The reaction mixture was cooled to O0C, and slowly basified with sat. Na2Cθ3(aq). The organic layer was separated and dried with Na2SO4, before evaporating, iv) The crude product was re-dissolved in DCM (10ml) and cooled to O0C. To this solution was added, N,N-diethylaminopropylsulfonyl chloride. Hydrochloride
(1.3eq), followed by DIPEA (3.0eq) and stirred overnight whilst allowing to warm to ambient temperatures. The reaction mixture was then washed with brine (2x1 OmI), dried over Na2SO4, and evaporated. The crude product was purified by silica gel column chromatography, eluting with a mixture of MeOH/DCM (typically 5/95).
Vinyl sulfonamide formation (6) iii) As described above v) As described for iv) (5) except 2-chloroethylsulfonyl chloride is to be used. Purification by silica gel column chromatography, eluting with EtOAc/Heptanes
(typically 1/9)
Diethylamine addition (7)
vi) N,N-Diethylamine (2ml) was added to (6) and stirred at ambient temperatures overnight. This was then evaporated to dryness in vacuo. The crude product was purified by silica gel column chromatography, ehiting with a mixture of MeOH/DCM (typically 5/95).
Preparation of N,N-3-Diethylaminopropanesulfonyl chloride hyrochloride 10.
Figure imgf000131_0001
Diethylamine (10ml, 96.78mmol) was added slowly to the 1,3 Propane sultone (3.94g, 32.26mmol) whilst stirring at O0C. This was allowed to stir overnight whilst warming to ambient temperatures. The reaction mixture was then diluted with MeOH (100ml) followed by the addition of Ambersep900 resin (3Og, 75mmol) and shaken for 2hours. The resin was washed, on a glass sinter with 30ml aliquots of MeOH (washings discarded). The resin was then treated with 2M HCl(aq) (50ml) for 30mins, and then filtered. The resin was washed with 2M HCl(aq; (2 x 50ml). The combined acidic solution was evaporated in vacuo to give the crude product N,N-3-diethylaminopropane sulfonic acid hydrochloride (8) as a white solid.
Thionyl chloride (50ml, 685mmol) was added to N,N-3-diethylaminopropane sulfonic acid hydrochloride (8) and heated to reflux whilst stirring for δhours. The solution was allowed to cool to ambient temperatures and then evaporated in vacuo to give N,N-3-diethylaminopropanesulfonyl chloride hyrochloride (9) as a grey/white, very hydroscopic solid (7.9g, 92%). The product was stored under a nitrogen atmosphere.
Example 4: Synthesis of (RVtert-butyl 3-phenyl-l-(3-(pyridin-2-vD-lH-L2,4-triazol- 5-yl)propγlcarbamate
Stage-1
Figure imgf000131_0002
2-Cyano pyridine (500mg, 1.923mmol) and Hydrazine hydrate (252mg, 1.923mmol) were mixed together in EtOH (3ml). The clear solution was left at 15 0C (in refrigerator) for 18h (Monitored by TLC). Ethanol was concentrated under reduced pressure and then the reaction mixture was diluted with diethyl ether (50ml) and water. The diethyl ether layer was separated, washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to l/5fh Volume. The ethereal solution was cooled to 0 0C and the obtained solid precipitate was collected over the filtration and dried under vacuum, which gave the 2-pyridine Amidrazone (360mg).
Stage-2:
Figure imgf000132_0001
To a solution of the Boc protected Homophenyl alanine (1.Ig, 3.012mmol) and TEA (370mg, 3.012mmol) in dry THF (10ml) at -5 0C was added ethyl chloro formate (400mg) drop wise and stirred the reaction mixture for 30min at the same temperature. TLC shows the formation of activated ester. After completion of the reaction, mixture was filtered through celite (to remove the Triethyl ammonium hydrochloride) and to the filtrate was added the Amidrazone (350mg) and stirred at RT for 5h under N2 atmosphere. Progress of the reaction was monitored by TLC analysis. After completion of the reaction, water was added and extracted the mixture with ethyl acetate. The organic layer was washed with water, brine, dried over Na2SO4, filtered and concentrated to provide the Uncyclized intermediate (500mg), which was taken to next step without purification. Stage-3:
Figure imgf000133_0001
The Uncyclized crude intermediate (500mg) was taken in xylenes (mixture of xylenes) and heated to reflux (175 0C) under Dean-Stark conditions for 6h. Progress of the reaction was monitored by TLC analysis, after completion of the reaction, xylenes were distilled off under reduced pressure and the crude residue was purified by flash column chromatography using silica gel to provide 320mg of the required triazole, confirmed by MS and NMR.
Example 5: Synthesis of (R)-3-(diethvIaminoVN-ri-(3-(4-fluoroOhenylVlH-1.2.4- triazo]-5-γl)-3-phenylpropyl)propane-l-sulfonamide
Figure imgf000133_0002
Figure imgf000133_0003
Figure imgf000133_0004
Synthesis of 4-fluorobenzothioamide: To a solution of 4-fluoro benzonitrile (1) (2.5g, 20.1mmol) in THF: water (5ml: 15ml) was added diethyl dithiophosphate (2) (10.5g, 60.3mmol) at room temperature and continued the reaction at 80 oC for 8 h. Progress of the reaction was monitored by TLC analysis. To the reaction mixture was added water (25ml) and the compound was extracted with ethyl acetate (2 x 25ml). The organic layer was washed with brine, dried over sodium sulfate, filtered, concentrated under reduced pressure to give the crude thioamide-3 (2.8g), taken to next step without further purification.
Synthesis of methyl 4-fluorobenzimidothioate: To a solution of Thioamide (3) (2.8g, 18.2mmol) in acetonitrile (15ml) was added methyl iodide (11.7ml, 90.3mmol) at room temperature and continued the reaction at 80 oC for 1 h. Progress of the reaction was monitored by TLC analysis. The solvent from the reaction mixture was removed under reduced pressure. The obtained crude residue was purified by crystallization using diethyl ether, which gave the thiomethyl derivative- 4 (2g). IH NMR-500MHZ (DMSO-D6)δ 11.62 (IH, bm), 8.01 (2H, m), 7.47 (2H, m), 2.92 (3H, s); MS 170 (M+, 100%).
Synthesis of (R)-tert-butyl l-hydrazinyl-l-oxo-4-phenylbutan-2-ylcarbamate:
Boc-HomoPhe-methylester (5) (3g, 10.2mmol) was taken in hydrazine hydrate (99% solution (10ml) at room temperature and continued the reaction at 80 oC for 3 h.
Progress of the reaction was monitored by TLC analysis. The reaction was diluted with water and extracted with dichloromethane (2 x 25ml). The organic layer was washed with water, brine, dried over sodium sulfate, filtered, concentrated under pressure to provide compound-6 (2.8g) as white solid. IH NMR-200MHZ (CDCl3)S 7.61 (IH, bs), 7.37-7.21 (5H, m), 5.14 (IH, m), 4.04 (IH, m), 3.81 (2H, bs), 2.67 (2H, t, J = 5.2Hz),
2.21-1.84 (2H, m), 1.42 (9H, s); MS 294 (M+, 100%), 194 (M-Boc, 75%).
Synthesis of (R)-tert-butyϊ l-(3-(4-fluorophenyl)-lH-l,2,4-triazol-5-yI)-3- phenylpropylcarbamate: To a solution of compound-6 (2g, 6.82mmol) in ethanol (20ml) were added compound-4 (2.88g, 17.05mmol) followed by DIPEA (6.5ml, 17.05mmol) at 0 0C and continued the reaction at 80 0C for 5 h. Progress of the reaction was monitored by TLC analysis. Ethanol was removed from the reaction mixture under reduced pressure and the obtained residue was diluted with water (10ml) and ethyl acetate (25ml). The organic layer was separated, washed with water, brine, dried over sodium sulfate, filtered, concentrated under pressure to provide compound-7 (2.3g). IH NMR-200MHZ (CDCl3)δ 12.01 (IH, bm), 8.06 (2H, m), 7.28-7.12 (7H, m), 5.24 (IH, m), 4.82 (IH, m), 2.73 (2H, t, J= 5.2Hz), 2.42-2.18 (2H, m), 1.44 (9H, s); MS 396 (M+, 100%), 296 (M-Boc, 75%).
Synthesis of 3-(diethylamino)propane-l-sulfonyl chloride:
To a solution of 1,3-propanesultone (5g, 40.98mmol) in dichloromethane (20ml) was added diethyl amine (15ml, 204.9mmol) drop wise at 10 0C and continued the reaction for Ih. Progress of the reaction was monitored by TLC analysis. Dichloromethane was removed from the reaction mixture under reduced pressure and the obtained residue was crystallized from diethyl ether to produce the sulfonic acid compound as solid (4.9g) IH NMR-200MHZ (CDCl3)δ 9.44(1H, bm), 3.16 (6H, m), 2.63 (2H, t, J= 5.2Hz), 1.96 (2H, m), 1.20 (6H, t, J= 5.2Hz); MS 195 (M+, 100%).
The above-obtained sulfonic acid (4.9g) was taken in Thionyl chloride (40ml) and the reaction mixture was continued stirring at 80 0C for 5h. Progress of the reaction was monitored by TLC analysis. Excess Thionyl chloride was removed under reduced pressure and the obtained residue was thoroughly azeotroped using toluene (3 x50ml) under nitrogen atmosphere. The crude residue was washed thoroughly with diethyl ether (3 x 10ml) followed by hexane (3 x 10ml) to give pure compound-A, which was used in the next reaction without further purification.
Synthesis of (R)-l-(3-(4-fluorophenyl)-lH-l,2,4-triazol-5-yl)-3-phenylpropan-l- amine hydrochloride: Compound-7 (2.3g, 5.808mmol) was taken in methanolic HCl (20% solution,
20ml) at 0 0C and continued the reaction at room temperature for 3 h. Progress of the reaction was monitored by TLC analysis. Solvent was removed from the reaction mixture under reduced pressure and the obtained solid was thoroughly washed with diethyl ether to produce the compound-8-HCl (2g) IH NMR-200MHZ (DMSO-D6)S 8.71(1H, bm), 8.12 (2H, m), 7.43-7.16 (7H, m), 4.42 (IH, m), 2.65 (2H, t, J = 5.2Hz), 2.22 (2H, m); MS 297 (M+, 80%).
Synethesis of (R)-3-(diethylamino)-N-(l-(3-(4-fluorophenyl)-lH-l,2,4-triazol-5-yl)- 3-phenylpropyl)propane-l-sulfonamide: To a 0 0C cooled solution of Compound-8 (250mg, 0.753mmol) in dichloromethane (5ml) was added triethylamine (0.5ml, 4.518mmol). To the reaction mixture was added a solution of compound- A (240mg, 1.129mmol) in dichloromethane (2.5ml) drop wise and the reaction mixture was continued stirring for Ih. Progress of the reaction was monitored by TLC analysis. The reaction mixture was diluted with dichloromethane (20ml), washed with saturated sodium bicarbonate solution, water followed by brine solution. The organic layer was dried over sodium sulfate, filtered, concentrated under reduced pressure to provide the crude material. The crude residue was purified by flash column chromatography on silica gel using methanol: dichloromethane (4:96) as eluent. The obtained compound was taken in dichloromethane and washed with water. The organic layer was dried over sodium sulfate, filtered, concentrated under reduced pressure to provide pure compound as thick brown (155mg) IH NMR-200MHz (CD3OD)δ 8.08 (2H, m), 7.37-7.16 (7H, m), 4.63 (IH, t, J = 5.2Hz), 2.98 (8H, m), 2.35 (2H, m), 1.95 (2H, m), 1.05 (6H, t, J = 5.2Hz); MS 473.8 (M+, 100%).
Example 6: Reduced ghrelin-induced food intake in mice dosed ip with compound
57
Ad libidum low fat chow-fed male C57bl/6 mice were acclimated to individual housing for >5 days prior to the experiment. On the morning of the experiment food was removed from the cages and animals were then dosed ip (n=8/group) with either vehicle or ghrelin followed one minute later by a second ip injection containing either vehicle or one of several doses of compound (1, 3, 10, 30mpk ip). Immediately after the second dosing food was returned to the cages and food intake was measured 1, 2, 4 and 6 hours later. The results are provided in Figures 1 and 2, demonstrating reduced food intake in mice dosed with compound 57 and compound 94 respectively relative to mice dosed with vehicle.
Example 7: Exemplary compounds were orally bioavailable
Male, diet-induced obese, ad libidum fed, C57bl/6 mice were used for these preliminary PK studies. They were dosed (n=3 mice per data point) either IV or PO. Mice ere sacrificed and the blood/brains collected at the following times: IV samples were taken at 5, 15, 30, 60, 120, 240, 480 minutes after dosing; PO samples were taken at 15, 30, 60, 120, 240, 480 minutes after dosing. Plasma and brain samples were evaluated for the presence of compound. Oral bioavailability (%F) was determined by comparing the plasma AUQv and AUCpo The brain/plasma ratio was determined by comparing the brain AUC and plasma AUC values. The results, provided in Table 2 below, demonstrate that the sample compounds 57 and 94 are orally bioavailable.
Figure imgf000137_0001
Example 8: 28 days oral dosing of compounds 57 and 94 caused a decrease in body weight with no apparent effect on food intake
Group housed, male, C57bl/6 mice received high fat diet and water ad lib to establish diet-induced obesity, and for the duration of the experiment. After acclimating to individual housing for at least 5 days, mice were then acclimated to bid oral gavage with vehicle for 7 days. Once acclimated, mice were culled into equal groups (n>10/group) according to body weight and compound dosing began. Mice received either vehicle or compound (bid, po) for periods up to 28 days, during which body weight and food intake were measured daily. Representative cohorts (based on BW) for each group were removed at 7, 14, 21 or 28 days for glucose tolerance tests and tissue analysis. As shown in Figure 3a and Figure 3b, administration of compound 57 and compound 94 provided a decrease in body weight. No apparent effect on food intake was observed.
Example 9: 7 to 28 days oral dosing of compounds 57 and 95 improved insulin sensitivity
On the night prior to the GTT, mice (DIO) within each dose group (n=8/group) had their body weight measured and were then fasted overnight for 16 hours prior to the GTT. On the morning of the GTT a 40DL blood sample was removed from the tail for baseline fasted blood glucose and plasma insulin measurements. Mice then received their normal dose of compound. One hour later mice were dosed with glucose (lmg/Kg, either IP or PO) and further tail bleeds (40DL) were taken at 15, 30, 60 and 120 minutes after the glucose dose for blood glucose and plasma insulin measurements. Mice were sacrificed following the final sample time. Baseline fasted blood glucose levels are provided in Figure 4a for compound 57 and in Figure 4d for compound 94. Figure 4b shows the data demonstrating improved insulin sensitivity of mice dosed with compound 57 at 7 days, whereas Figure 4e shows the data demonstrating improved insulin sensitivity of mice dosed with compound 94 at 7 days. Figure 4c shows the data demonstrating improved insulin sensitivity of mice dosed with compound 57 at 21 days. Figure 4f shows the data demonstrating improved insulin sensitivity of mice dosed with compound 94 at 28 days.
ExamplelO: 28 day dosing of compound 57 and compound 94 results in improved plasma HbAIc and cholesterol levels
Mice are sacrificed by CO2 asphyxiation and terminal blood samples were collected via cardiac puncture. For HbAIc, whole blood is analyzed using a DCA2000+ glycated hemoglobin analyzer (Bayer Healthcare, Indianapolis, IN). Triglycerides, total cholesterol, and HDL-cholesterol were all measured using a Cholestech LDX lipid profile analyzer (Cholestech, Hayward, CA). As shown in Figure 6 and Figure 7, both plasma
HbAIc and cholesterol levels were improved at 28 days in animals treated with compound 57 and compound 94 versus animals treated with vehicle alone. Example U: 14 day oral dosing with compound 57 improves fasting blood glucose in db/db mice
Mice that had been acclimated to individual housing and oral gavage dosing were fasted for 16 hours overnight. The following morning a blood sample was removed from the tail for baseline fasted blood glucose measurements. This process was repeated after 14 days oral bid dosing with compound 57. Whereas fasted blood glucose levels continued to rise in vehicle treated animals, there was no change in mice treated with the ghrelin antagonist. Thus, compound treatment blocked the development of hyperglycemia in this model of type 2 diabetes. Figure 7 illustrates the improvement in fasting blood glucose levels of mice treated with compound 57.
A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other embodiments are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A pharmaceutical composition comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
2. The pharmaceutical composition of claim 1, wherein the additional therapeutic agent is an insulin preparation, an oral hypoglycemic, a peroxisome proliferator-activated receptor gamma agonist, a nitrate, a nitrite, a β-blocker, a calcium channel antagonist, a peripheral vasodilator, a cinchona alkaloid, an amide, a quaternary ammonium salt, a diidobenzyloxyethylamine, a hydantoin, a diuretic, a vasodilator, a peripheral sympatholytic, a central α2 -sympathomimetic, an angiotensin-converting enzyme inhibitor, an angiotensin II-receptor antagonist, a HMG CoA reductase inhibitor, a fϊbratc, an antiarrhythmic, a cardiac glycoside, a heparin, a coumarin derivative, a histonc deacetylase (HDAC) inhibitor, a sirtuin (SIRT) inhibitor, a xanthine oxidase inhibitor, an alkylating agent, an antitumor antibiotic, an antimetabolite, a mitotic inhibitor, a topoisomerase inhibitor, an enzyme, a protein tyrosine kinase inhibitor, a hormonal agent, an aromatase inhibitor, a LHRH analogue, a biological agent, a herbal remedy, an oncolytic adenovirus, a benzodiazepan, an opioid-receptor antagonist, an antidepressant, an analgesic, a hyaluronic acid derivative, or an amphetamine.
3. A solid dosage form comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI, and an additional therapeutic agent.
5. The solid dosage form of claim 3, wherein the additional therapeutic agent is an insulin preparation, an oral hypoglycemic, a peroxisome proliferator-activated receptor gamma agonist, a nitrate, a nitrite, a β-blocker, a calcium channel antagonist, a peripheral vasodilator, a cinchona alkaloid, an amide, a quaternary ammonium salt, a diidobenzyloxyethylamine, a hydantoin, a diuretic, a vasodilator, a peripheral sympatholytic, a central α2 -sympathomimetic, an angiotensin-converting enzyme inhibitor, an angiotensin II-receptor antagonist, a HMG CoA reductase inhibitor, a fibrate, an antiarrhythmic, a cardiac glycoside, a heparin, a coumarin derivative, a histone deacetylase (HDAC) inhibitor, a sirtuin (SIRT) inhibitor, a xanthine oxidase inhibitor, an alkylating agent, an antitumor antibiotic, an antimetabolite, a mitotic inhibitor, a topoisomerase inhibitor, an enzyme, a protein tyrosine kinase inhibitor, a hormonal agent, an aromatase inhibitor, a LHRH analogue, a biological agent, a herbal remedy, an oncolytic adenovirus, a benzodiazepan, an opioid-receptor antagonist, an antidepressant, an analgesic, a hyaluronic acid derivative, or an amphetamine.
6. The solid dosage form of claim 3, comprising a tablet or capsule.
7. The solid dosage form of claim 6, further comprising an enteric coating.
8. A method of treating a disorder in a subject, the method comprising administering to the subject a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an additional therapeutic agent.
9. The method of claim 8, wherein the ghrelin antagonist is co-administered with the additional therapeutic agent.
10. The method of claim 8, wherein the ghrelin antagonist and the additional therapeutic agent are administered in a single dosage form.
11. The method of claim 8, wherein the ghrelin antagonist and the additional therapeutic agent are provided in amounts so as to provide a synergistic effect.
12. The method of claim 8, wherein the ghrelin antagonist and the additional therapeutic agent are administered one time daily.
13. The method of claim 8, wherein the ghrelin antagonist and the additional therapeutic agent are administered twice daily.
14. The method of claim 8, wherein the ghrelin antagonist and the additional therapeutic agent are administered three times daily.
15. The method of claim 8, wherein the ghrelin antagonist and the additional therapeutic agent are administered within 1 hour before or two hours after a meal.
16. The method of claim 8, wherein the ghrelin antagonist and the additional therapeutic agent are administered without regard to food.
17. The method of claim 8, wherein the disorder is obesity.
18. The method of claim 8, wherein the disorder is a combination of disorders.
19. The method of claim 18, wherein the combination of disorders is obesity and an additional disorder.
20. The method of claim 8, wherein the additional therapeutic agent is an insulin preparation, an oral hypoglycemic, a peroxisome proliferator-activated receptor gamma agonist, a nitrate, a nitrite, a β-blocker, a calcium channel antagonist, a peripheral vasodilator, a cinchona alkaloid, an amide, a quaternary ammonium salt, a diidobenzyloxyethylamine, a hydantoin, a diuretic, a vasodilator, a peripheral sympatholytic, a central ^-sympathomimetic, an angiotensin-converting enzyme inhibitor, an angiotensin II-receptor antagonist, a HMG CoA reductase inhibitor, a fibrate, an antiarrhythmic, a cardiac glycoside, a heparin, a coumarin derivative, a histone deacetylase (HDAC) inhibitor, a sirtuin (SIRT) inhibitor, a xanthine oxidase inhibitor, an alkylating agent, an antitumor antibiotic, an antimetabolite, a mitotic inhibitor, a topoisomerase inhibitor, an enzyme, a protein tyrosine kinase inhibitor, a hormonal agent, an aromatase inhibitor, a LHRH analogue, a biological agent, a herbal remedy, an oncolytic adenovirus, a benzodiazepan, an opioid-receptor antagonist, an antidepressant, an analgesic, a hyaluronic acid derivative, or an amphetamine.
21. The method of claim 20, wherein the insulin preparation is a short acting insulin, regular insulin, semilente insulin, intermediate acting insulin, lente insulin, or long acting insulin.
22. The method of claim 20, wherein the oral hypoglycemic is a sulfonylurea, a meglitinide, α-glucosidase inhibitor, or an insulin sensitizer.
22. The method of claim 22, wherein the sulfonylurea is tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, or glimepiride.
23. The method of claim 22, wherein the meglitinide is mitiglinide, repaglinide or nateglinide.
24. The method of claim 22, wherein the α-glucosidase inhibitor is acarbose, miglitol, voglibose (N-l(l ,3-dihydroxy-2-propyl) valiolamine, and N-substituted derivatives thereof, and N-substitutcd pscudo-amino sugars.
25. The method of claim 22, wherein the insulin sensitizer is a biguanide, or a thiazolidinedione.
26. The method of claim 25, wherein the biguanide is metformin.
27. The method of claim 25, wherein the thiazolidinedione is pioglitazone or rosiglitazone.
28. The method of claim 20, wherein the peroxisome proliferator-activated receptor gamma agonist is a thiazolidinedione.
29. The method of claim 20, wherein the nitrate is nitroglycerin, isorbide, isorbide-5 -mononitrate, or erythrityl tetranitrate.
30. The method of claim 20, wherein the nitrite is amyl nitrate.
31. The method of claim 20, wherein the β-blocker is propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, cartcolol, oxprcnolol, pcnbutolol, lcvatol, mcdroxalol, bucindolol, levobunolol, metipranolol, bisoprolol, nebivolol, betaxolol, celiprolol, sotalol, or propafenone.
32. The method of claim 20, wherein the calcium channel antagonist is amlodipine, arandipine, bamidipine, bencyclane, benidipine, bepridil, cilnidipine, cinnarizine, clentiazem, diltiazem, efonidipine, elgodipine, etafenone, flunarizine, fϊlodepine, galopamil, isradipine, lacidipine, lercanidipine, lidoflazine, lomerizine, manidipine, mibefradil, nicardipine, nifedipine, nilvadepine, nimodipine, nisoldipine, nitrendipine, penylamine, perhexiline, phendilin, semotiadil, terodiline, or verapamil.
33. The method of claim 20, wherein the peripheral vasodilator is dipyridamole, nylidrin hydrochloride, or isoxsuprine hydrochloride.
34. The method of claim 20, wherein the cinchona alkaloids is quinidine or quinine.
35. The method of claim 20, wherein the amide is procainamide, flecainide, lidocaine, bupivicaine, ropivicaine, or dispyramide.
36. The method of claim 20, wherein the diidobenzyloxyethylamine is amiodarone.
37. The method of claim 20, wherein the hydantoin is phenytoin.
38. The method of claim 20, wherein the diuretic is a thiazide diuretic, an osmotic diuretic, a carbonic anhydrase inhibitor, a benzothiadiazide diuretic, a loop diuretic, or a potassium sparing diuretic.
39. The method of claim 38, wherein the thiazide diuretic is bendrofluazide, bendrofiumethiazide, benzthiazide, benzylhydrochlorothiazide, chlorothiazide, chlorthalidone, cyclopenthiazide, diucardin, diuril, enduron, esidrix, exna, HCTZ, hydrochlorothiazide, hydrodiuril, hydroflumethiazide, bendrofiumethiazide, hydromox, hygroton, indapamide, lozol, methylchlorthiazide, metolazone, mykrox, naqua, naturetin, oretic, penfiutizide, polythiazide, quinethazone, renese, trichlormethiazide, xipamide, or zaroxolyn.
40. The method of claim 38, wherein the osmotic diuretic is mannitol, urea, glycerin, or isorbide.
41. The method of claim 38, wherein the carbonic anhydrase inhibitor is acetazolamide.
42. The method of claim 38, wherein the benzothiadiazide diuretic is or chlorothiazide.
43. The method of claim 38, wherein the loop diuretic is azosemide, furosemide, bumetanide, ethacrynic acid, piretanide, or torasemide.
44. The method of claim 38, wherein the potassium sparing diuretic is triamterene, amiloride, spironolactone, or eplerenone.
45. The method of claim 20, wherein the vasodilator is bencyclane, cinnarizine, citicoline, cyclandate, cyclonicate, diazoxide, ebumamonine, flunarizine, hydralazine, ibudilast, ifenprodil, lomerizine, minoxidil, naphlole, nikamate, nimodipine, nitroprusside, nofedoline, nosergoline, papaverine, pentifylline, pentoxifylline, phenoxezyl, prostacyclin derivatives, vichizyl, vincamin, or vinpocetine.
46. The method of claim 20, wherein the peripheral sympatholytic is acebutol, atenolol, betaxolol, bisoprolol, carteolol, carvediolol, doxazosin, guanadrel, guanethidine, labetalol, metoprolol, nadolol, penbutolol, pindolol, prazosin, propanolol, reserpine, terazosin, or timolol.
47. The method of claim 20, wherein the central α2-sympathomimetic is clonidine, guanabenz, guanfacine, or methyldopa.
48. The method of claim 20, wherein the angiotensin-converting enzyme inhibitor is alacepril, benazepril, captopril, ceranopril, cilazopril, delapril, enalapril, fosinopril, imidapril, lisinopril, mobertpril, moexipril, pentopril, perindopril, quinapril, ramipril, randolapril, spirapril, temocapril, trandolapril, or zofenopril.
49. The method of claim 20, wherein the angiotensin II-receptor antagonist is candesartan, eprosartan, ibersartan, losartan, telmisartan, or valsartan.
50. The method of claim 20, wherein the HMG CoA reductase inhibitor is livostatin, simvastatin, pravastatin, fluvastatin, atorvastatin, or cerivastatin.
51. The method of claim 20, wherein the fibrate is nicotinic acid, bezafibrate, clinofibrate, clofibrate, colestimide, fenofibrate, gemfϊbrizil, probucol, or simfibrate.
52. The method of claim 20, wherein the antiarrhythmic is tocainide, encainide, phenytoin, flecainide, tocainide, propafenone, bretylium, sotalol, ibutilide, dofetilide, bepridil, moricizine, propanolol, esmolol, artilide, clofϊlium, azimilide, dronedarone, ersentilide, ibutilide, tedisamil, trecetilide, verapamil, diltaizem, digitalis, adenosine, nickel chloride, procainaltide, lignocaine, or magnesium ions.
53. The method of claim 20, wherein the cardiac glycoside is allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, a xanthine formulation, a catecholamine formulation, denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, or colforsin daropate.
54. The method of claim 53, wherein the xanthine formulation is aminophylline, choline theophylline, diprophylline, or proxphylline.
55. The method of claim 53, wherein the catecholamine formulation is dopamine, dobutamine, or docarpamine.
56. The method of claim 20, wherein the heparin agent is heparin sodium, heparin potassium, a low-molecular weight heparin fragment, or a low-molecular weight heparinoid.
57. The method of claim 56, wherein the low-molecular weight heparin fragment is enoxaparin, dalteparin, tinzaparin, or ardeparin.
58. The method of claim 56, wherein the low-molecular weight heparinoid is danaparoid.
59. The method of claim 20, wherein the coumarin derivative is warfarin or dicumarol.
60. The method of claim 20, wherein the histone deacetylase inhibitor is a class I and/or a class II histone deacetylase inhibitor.
61. The method of claim 60, wherein the inhibitor of the class I and/or class II histone deacetylase is a small molecular weight carboxylate.
62. The method of claim 60, wherein the inhibitor of the class I and/or class II histone deacetylase is a hydroxamic acid.
63. The method of claim 60, wherein the inhibitor of the class I and/or class II histone deacetylase is a benzamide.
64. The method of claim 60, wherein the inhibitor of the class I and/or class II histone deacetylase is an epoxyketone.
65. The method of claim 60, wherein the inhibitor of the class I and/or class II histone deacetylase is a cyclic peptide.
66. The method of claim 60, wherein the inhibitor of the class I and/or class II histone deacetylase is a hybrid molecule.
67. The method of claim 60, wherein the inhibitor of the class I and/or class II histone deacetylase is Suberoylanilide Hydroxamic Acid (SAHA (e.g., MK0683, vorinostat) and other hydroxamic acids), Suberoyl bis-hydroxamic Acid, BML-210, Depudecin (e.g., (-)-Depudecin), HC Toxin, Nullscript (4-(l,3-Dioxo-lH,3H- benzo[de]isoquinolin-2-yl)-N-hydroxybutanamide), Phenylbutyrate (e.g., sodium phenylbutyrate) and Valproic Acid (and other short chain fatty acids), Scriptaid, Splitomicin, Suramin Sodium, Trichostatin A (TSA), APHA Compound 8, Apicidin, Sodium Butyrate, pivaloyloxymethyl butyrate (Pivanex, AN-9), Trapoxin B, Chlamydocin, Depsipeptide (also known as FR901228 or FK228), benzamides (e.g., CI- 994 (i.e., N-acetyl dinaline) and MS-27-275), MGCD0103, NVP-LAQ-824, CBHA (m- carboxycinnaminic acid bishydroxamic acid), JNJ16241199, Tubacin, A-161906, proxamide, oxamflatin, 3-Cl-UCHA (i.e., 6-(3-chlorophenylureido)caproic hydroxamic acid), AOE (2-amino-8-oxo-9,10-epoxydecanoic acid), CHAP31 or CHAP 50.
68. The method of claim 20, wherein the SIRT inhibitor is a 2-anilinobenzamide or 2-chloro-5,6,7,8,9,10-hexahydrocyclohepta[b]indole-6, 2-chloro-5,6,7,8,9,10- hexahydrocyclohepta[b]indole-6-carboxylate, or 2-chloro-5,6,7,8,9, 10- hexahydrocyclohepta[b]indole-6-carboxamide, or 6-Chloro-2,3 ,4,9-tetrahydro- 1 H- carbazole-1-carboxylic acid amide.
69. The method of claim 68, wherein the 2-anilinobenzamide is nicotinamide, carba-NAD+, EX-527, sirtinol, para-sirtinol, JFD00244, splitomicin, HR73, or cambinol.
70. The method of claim 20, wherein the xanthine oxidase inhibitor is allopurinol.
71. The method of claim 20, wherein the alkylating agent is altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, or thiotepa.
72. The method of claim 20, wherein the antitumor antibiotic is bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitoxantrone, or valrubicin.
73. The method of claim 20, wherein the antimetabolite is capecitabine, cladribine, cytarabine, fludarabine, floxuridine, 5-fluorouracil, gemicabine, hydroxyurea, 6-mercaptopurine, methotrexate, pentostatin, or 6-thioguanine.
74. The method of claim 20, wherein the mitotic inhibitor is docetaxel, paclitaxel, vinblastine, vincristine, or vinorelbine.
75. The method of claim 20, wherein the topoisomerase inhibitor is etoposide, irinotecan, teniposide, or topotecan.
76. The method of claim 20, wherein the enzyme is asparaginase or pagasparaginase.
77. The method of claim 20, wherein the protein tyrosine kinase inhibitor is imatinib mesylate.
78. The method of claim 20, wherein the hormonal agent is an adrenocorticoid, an estrogen, a progestin, an antiestrogen, an androgen, or an antiandrogen.
79. The method of claim 78, wherein the adrenocorticoid is dexamethasone, methylprednisolone, or prednisone.
80. The method of claim 78, wherein the estrogen is diethylstilbestrol or estradiol.
81. The method of claim 78, wherein the progestin is medroxyprogesterone or megesterol acetate.
82. The method of claim 78, wherein the antiestrogen is fulvestrant, tamoxifen, or toremifene.
83. The method of claim 78, wherein the androgen is testosterone, methyltestosterone, or fluoxymesterone.
84. The method of claim 78, wherein the antiandrogen is bicalutamide, flutamide, or nilutamide.
85. The method of claim 20, wherein the aromatase inhibitor is aminoglutethimide, anastrozole, exemestane, or letrozole.
86. The method of claim 20, wherein the LHRH analogue is leuprolide or goserelin.
87. The method of claim 20, wherein the biological agent is interferon-α 2a and 2b, inter leukin-2, interleukin- 11 , the growth factors filgrastim, pegfϊlgrastim, sargramostim, epoetin alfa, darbepoetin alfa, or a monoclonal antibody.
88. The method of claim 20, wherein the benzodiazepine is diazepam, lorazcpam, oxazepam, ativan, serax, temazepam, clonazepam, valium, and xanax.
89. The method of claim 20, wherein the opioid-receptor antagonist is tramadol or naltrexone.
90. The method of claim 20, wherein the antidepressant is a selective serotonin reuptake inhibitor, a serotonin and norepinephrine reuptake inhibitor, a tetracyclic antidepressant, a norepinephrine and dopamine reuptake inhibitor, a combined reuptake inhibitor and receptor blocker, a tricyclic antidepressant, or a monoamine oxidase inhibitor.
91. The method of claim 90, wherein the selective serotonin reuptake inhibitor is citalopram, escitalopram, fluoxetine, paroxetine, or sertraline.
92. The method of claim 90, wherein the serotonin and norepinephrine reuptake inhibitor is duloxetine or venlafaxine.
93. The method of claim 90, wherein the tetracyclic antidepressant is mirtazapine.
94. The method of claim 90, wherein the combined reuptake inhibitor and receptor blocker is trazodone, nefazodone, or maprotiline.
95. The method of claim 90, wherein the tricyclic antidepressant is amitriptyline, amoxapine, desipramine, doxepin, imipramine, nortriptyline, protriptyline, or trimipramine.
96. The method of claim 90, wherein the monoamine oxidase inhibitor is phenelzine, ranylcypromine, isocarboxazid, or selegiline.
97. The method of claim 20, wherein the analgesic is paracetamol, a nonsteroidal anti-inflammatory drug, a cyclooxygenase-2 inhibitor, an opiate, a corticosteroid, or a topical analgesic.
98. The method of claim 97, wherein the paracetamol is acetaminophen or indomethacin.
99. The method of claim 97, wherein the non-steroidal anti-inflammatory drugs comprise, aspirin, ibuprofen, salsalate, magnesium salicylate, or a COX-2 selective inhibitor.
100. The composition of claim 99, wherein the COX-2 selective inhibitor is celecoxib, refecoxib, or valdecoxib.
101. The method of claim 97, wherein the opiate is codeine, oxycodone, hydrocodone, diamorphine, pethidine, tramadol, or buprenorphine.
103. The method of claim 97, wherein the corticosteroid is prednisone, prednisolone, cortisone, or hydrocortisone.
104. The method of claim 97, wherein the topical analgesic is capsaicin.
105. The method of claim 20, wherein the amphetamine is dextroamphetamine, benzedrine, ritalin, phenmetrazine, phendimetrazine, methamphetamine, diethylpropion, phentermine, sibrutamine, or orlistat.
106. The method of claim 5, wherein the disorder is obesity.
107. The method of claim 8, wherein the disorder is a combination of disorders.
108. The method of claim 107, wherein the combination of disorders is obesity and syndrome X.
109. The method of claim 108, wherein syndrome X is diabetes.
110. The method of claim 8, wherein the additional therapeutic agent is an antidiabetic agent.
111. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-diabetic agent.
112. The method of claim 111, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered in a single dosage form.
113. The method of claim 111, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are provided in amounts so as to provide a synergistic effect.
114. The method of claim 111, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered one time daily.
115. The method of claim 111, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered twice daily.
116. The method of claim 111, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered three times daily.
117. The method of claim 111, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered within 1 hour before or two hours after a meal.
118. The method of claim 111, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and the anti-diabetic agent are administered without regard to food.
119. A pharmaceutical composition comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-diabetic agent.
120. A solid dosage form comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-diabetic agent.
121. The solid dosage form of claim 120, comprising a tablet or capsule.
122. The solid dosage form of claim 121, further comprising an enteric coating.
123. The method of claim 111, wherein the anti-diabetic agent is insulin, tolbutamide, acetohexamide, chlorpropamide, glyburide, glipizide, glimepiride, repaglinide, nateglinide, acarbose, miglitol, metformin, pioglitazone, or rosiglitazone.
124. The method of claim 123, wherein the insulin is short acting insulin, regular insulin, semilente insulin, intermediate acting insulin, lente insulin, or long acting insulin.
125. The method of claim 107, wherein the combination of disorders is obesity and cardiovascular disease.
126. The method of claim 8 , wherein the additional therapeutic agent is a cardiovascular disease treating agent.
127. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with a cardiovascular disease treating agent.
128. The method of claim 127, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered in a single dosage form.
129. The method of claim 127, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are provided in amounts so as to provide a synergistic effect.
130. The method of claim 127, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered one time daily.
131. The method of claim 127, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered twice daily.
132. The method of claim 127, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered three times daily.
133. The method of claim 127, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered within 1 hour before or two hours after a meal.
134. The method of claim 127, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent are administered without regard to food.
135. A pharmaceutical composition comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent.
136. A solid dosage form comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a cardiovascular disease treating agent.
137. The solid dosage form of claim 136, comprising a tablet or capsule.
138. The solid dosage form of claim 137, further comprising an enteric coating.
139. The method of claim 127, wherein the cardiovascular treating agent is, allocar, corramedan, crystodigin, digitoxin, digoxin, methyldigoxin, lanoxin, purgoxin, aminophylline, choline theophylline, diprophylline, proxphylline, dopamine, dobutamine, docarpamine, denopamine, ubidecarenone, pimobendan, levosimendan, aminoethylsulfonic acid, vesnarinone, carperitide, colforsin daropate, amyl nitrite, nitroglycerin, propanolol, nadolol, timolol, pindolol, labetatol, atenolol, esmolol, acebutolol, metoprolol, carvedilol, bopindolpol, sandonorm, carteolol, oxprenolol, penbutolol, levatol, medroxalol, bucindolol, levobunolol, metipranolol, bisoprolol, nebivolol, betaxolol, celiprolol, sotalol, propafenone, verapamil, diltiazem, nicardipine, nifedipine, isradipine, amlodipine, felodipine, nisoldipine, nimodipine, bepridil, dipyridamole, nylidrin hydrochloride, isoxsuprine hydrochloride, quim'dine, procainamide, flecainide, lidocaine, bupivicaine, ropivicaine, dispyramide, lidocaine, mexiletine, bretylium, amiodarone, phenytoin, tocainide, encainide, phenytoin, flecainide, tocainide, propafenone, bretylium, sotalol, ibutilide, dofetilide, bepridil, moricizine, propanolol, esmolol, artilide, clofilium, azimilide, dronedarone, ersentilide, ibutilide, tedisamil, trecetilide, verapamil, diltaizem, digitalis, adenosine, nickel chloride, procainaltide, lignocaine, magnesium ions, bendrofluazide, bendroflumethiazide, benzthiazide, benzylhydrochlorothiazide, chlorothiazide, chlorthalidone, cyclopenthiazide, diucardin, diuril, enduron, esidrix, exna, HCTZ, hydrochlorothiazide, hydrodiuril, hydroflumethiazide, bendroflumethiazide, hydromox, hygroton, indapamide, lozol, methylchlorthiazide, metolazone, mykrox, naqua, naturetin, oretic, penflutizide, polythiazide, quinethazone, renese, trichlormethiazide, xipamide, zaroxolyn, mannitol, urea, glycerin, isorbide, acetazolamide, chlorothiazide, azosemide, furosemide, bumetanide, ethacrynic acid, piretanide, torasemide, triamterene, amiloride, spironolactone, eplerenone, bencyclane, cinnarizine, citicoline, cyclandate, cyclonicate, diazoxide, ebumamonine, flunarizine, hydralazine, ibudilast, ifenprodil, lomerizine, minoxidil, naphlole, nikamate, nimodipine, nitroprusside, nofedoline, nosergoline, papaverine, pentifylline, pentoxifylline, phenoxezyl, prostacyclin derivatives, vichizyl, vincamin, vinpocetine, acebutol, atenolol, betaxolol, bisoprolol, carteolol, carvediolol, doxazosin, guanadrel, guanethidine, labetalol, metoprolol, nadolol, penbutolol, pindolol, prazosin, propanolol, reserpine, terazosin, timolol, clonidine, guanabenz, guanfacine, methyldopa, amlodipine, arandipine, bamidipine, bencyclane, benidipine, bepridil, cilnidipine, cinnarizine, clentiazem, diltiazem, efonidipine, elgodipine, etafenone, flunarizine, fϊlodepine, galopamil, isradipine, lacidipine, lercanidipine, lidoflazine, lomerizine, manidipine, mibefradil, nicardipine, nifedipine, nilvadepine, nimodipine, nisoldipine, nitrendipine, penylamine, perhexiline, phendilin, semotiadil, terodiline, verapamil, candesartan, eprosartan, ibersartan, losartan, telmisartan, valsartan, alacepril, benazepril, captopril, ceranopril, cilazopril, delapril, enalapril, fosinopril, imidapril, lisinopril, mobertpril, moexipril, pentopril, perindopril, quinapril, ramipril, randolapril, spirapril, temocapril, trandolapril, zofenopril, cholestyramine chloride, colestipol hydrochloride, livostatin, simvastatin, pravastatin, fluvastatin, atorvastatin, cerivastatin, nicotinic acid, bezafϊbrate, clinofibrate, clofibrate, colestimide, fenofibrate, gemfϊbrizil, probucol, simfibrate, eicosapentaenoic acid, docosahexaenoic acid, heparin, heparin sodium, heparin potassium, enoxaparin, dalteparin, tinzaparin, ardeparin, danaparoid, lepirudin, warfarin, dicumarol, phenindione, aspirin, indomethacin, ticlopidine, cilostazol, clopidogrel, dipyridamole, tirofϊban, eptifibatide, anagrelide, Fab fragments of human monoclonal antibody to the GPIIb/IIIa receptor, alteplase, reteplase, tenecteplase, streptokinase, anistreplase, or urokinase.
140. The method of claim 107, wherein the combination of disorders is obesity and cancer.
141. The method of claim 8 , wherein the additional therapeutic agent is an anticancer agent.
142. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-cancer agent.
143. The method of claim 142, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered in a single dosage form.
144. The method of claim 142, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are provided in amounts so as to provide a synergistic effect.
145. The method of claim 142, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered one time daily.
146. The method of claim 142, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered twice daily.
147. The method of claim 142, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer treating agent are administered three times daily.
148. The method of claim 142, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered within 1 hour before or two hours after a meal.
149. The method of claim 142, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent are administered without regard to food.
150. A pharmaceutical composition comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent.
151. A solid dosage form comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-cancer agent.
152. The solid dosage form of claim 151, comprising a tablet or capsule.
153. The solid dosage form of claim 152, further comprising an enteric coating.
154. The method of claim 142, wherein the anticancer agent is, altretamine, busulfan, carmustine, chlorambucil, carboplatin, cisplatin, cyclophosphamide, dacarbazine, estramustine, ifosfamide, lomustine, mechlorethamine, melphalan, oxaliplatin, procarbazine, streptozocin, temozolamide, thiotepa, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, mitomycin C, mitoxantrone, valrubicin, capecitabine, cladribine, cytarabine, fludarabine, floxuridine, 5- fluorouracil, gemicabine, hydroxyurea, 6-mercaptopurine, methotrexate, pentostatin, 6- thioguanine, docetaxel, paclitaxel, vinblastine, vincristine, vinorelbine, etoposide, irinotecan, teniposide, topotecan, asparaginase, pagasparaginase, imatinib mesylate, dexamethasone, methylprednisolone, prednisone, diethylstilbestrol, estradiol, medroxyprogesterone, megesterol acetate, fulvestrant, tamoxifen, toremifene, aminoglutethimide, anastrozole, exemestane, letrozole, testosterone, methyltestosterone, fluoxymesterone, bicalutamide, flutamide, nilutamide, leuprolide, goserelin, interferon-α 2a and 2b, interleιιkin-2, interleukin-11, filgrastim, pegfilgrastim, sargramostim, epoetin alfa, darbepoetin alfa, monoclonal antibodies, or oncolytic adenoviruses.
155. The method of claim 107, wherein the combination of disorders is obesity and addiction.
156. The method of claim 8, wherein the additional therapeutic agent is an anti- addictive agent.
157. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula 1, 11, 111, IV, V, or Vl is co-administered with an anti-addictive agent.
158. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered in a single dosage form.
159. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are provided in amounts so as to provide a synergistic effect.
160. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered one time daily.
161. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered twice daily.
162. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive treating agent are administered three times daily.
163. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered within 1 hour before or two hours after a meal.
164. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent are administered without regard to food.
165. A pharmaceutical composition comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent.
166. A solid dosage form comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-addictive agent.
167. The solid dosage form of claim 166, comprising a tablet or capsule.
168. The solid dosage form of claim 167, further comprising an enteric coating.
169. The method of claim 157, wherein the anti-addictive agent is, nicotine replacement, diazepam, lorazepam, oxazepam, ativan, serax, temazepam, clonazepam, valium, xanax, propanolol, clonidine, bupropion hydrochloride, naltrexone, citalopram, fluoxetine, zimelidine, sertraline, paroxetine, disulfϊram, naltrexone, nalmefene, acamprosate, buspirone, or ondansetron.
170. The method of claim 107, wherein the combination of disorders is obesity and osteoarthritis.
171. The method of claim 8 , wherein the additional therapeutic agent is an anti- osteoarthritic agent.
171. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with an anti-osteoarthritic agent.
172. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered in a single dosage form.
173. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are provided in amounts so as to provide a synergistic effect.
174. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered one time daily.
175. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered twice daily.
176. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered three times daily.
177. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered within 1 hour before or two hours after a meal.
178. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent are administered without regard to food.
179. A pharmaceutical composition comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent.
180. A solid dosage form comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-osteoarthritic agent.
181. The solid dosage form of claim 180, comprising a tablet or capsule.
182. The solid dosage form of claim 181, further comprising an enteric coating.
183. The method of claim 171, wherein the antiosteoarthritic is, acetaminophen, salsalate, magnesium salicylate, aspirin, diclofenac, etodolac, ibuprofen, naproxen, nabumeton, sulindac, tolmetin, celecoxib, refecoxib, valdecoxib, tramadol, codeinek, oxycodone, capsaicin, corticosteroids, hyaluronic acid derivatives, glucosamine, chondroitin, or S-adenosyl-rnethionine.
184. The method of claim 8, wherein the additional therapeutic agent is a weight loss promoting agent.
185. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI is co-administered with a weight loss promoting agent.
186. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered in a single dosage form.
187. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are provided in amounts so as to provide a synergistic effect.
188. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered one time daily.
189. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered twice daily.
190. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered three times daily.
191. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered within 1 hour before or two hours after a meal.
192. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent are administered without regard to food.
193. A pharmaceutical composition comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent.
194. A solid dosage form comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and a weight loss promoting agent.
195. The solid dosage form of claim 194, comprising a tablet or capsule.
196. The solid dosage form of claim 195, further comprising an enteric coating.
197. The method of claim 185, wherein the weight loss promoting therapeutic agent is, dexfenfluramine, amphetamine, methamphetamine, phenmetrazine, phendimetrazine, diethylpropion, phentermine, sibutramine, or orlistat.
198. The method of claim 107, wherein the combination of disorders is obesity and gout.
199. The method of claim 8 , wherein the additional therapeutic agent is an anti- gout agent.
200. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered in a single dosage form.
201. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are provided in amounts so as to provide a synergistic effect.
202. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered one time daily.
203. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered twice daily.
204. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered three times daily.
205. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered within 1 hour before or two hours after a meal.
206. The method of claim 8, wherein the ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent are administered without regard to food.
207. A pharmaceutical composition comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent.
208. A solid dosage form comprising a ghrelin antagonist, for example a compound of formula I, II, III, IV, V, or VI and an anti-gout agent.
209. The solid dosage form of claim 208, comprising a tablet or capsule.
210. The solid dosage form of claim 209, further comprising an enteric coating.
211. The method of claim 200, wherein the anti-gout agent is, vorinostat, zolinza, SAHA, depsipeptide, MS-275, MGCD0103, PXDlOl, baceca, savicol, LBH589, PCI-
24781, or ITF2357.
PCT/US2008/063257 2007-05-09 2008-05-09 Ghrelin modulating compounds and combinations thereof WO2008141189A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US91705407P 2007-05-09 2007-05-09
US60/917,054 2007-05-09

Publications (1)

Publication Number Publication Date
WO2008141189A1 true WO2008141189A1 (en) 2008-11-20

Family

ID=40002615

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/063257 WO2008141189A1 (en) 2007-05-09 2008-05-09 Ghrelin modulating compounds and combinations thereof

Country Status (1)

Country Link
WO (1) WO2008141189A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20110007978A (en) * 2009-07-17 2011-01-25 한국생명공학연구원 A composition for preventing and treating bone disease comprising colforsin daropate
JP2012528890A (en) * 2009-06-05 2012-11-15 ティーエーユー・セラピューティクス・エルエルシー Combination methods for treating cancer or precancerous conditions
US8410167B2 (en) 2008-04-17 2013-04-02 Sanofi Use of dronedarone for the preparation of a medicament for use in the prevention of cardiovascular hospitalization or of mortality
WO2013175500A1 (en) * 2012-04-23 2013-11-28 Cadila Healthcare Limited Delazed release pharmaceutical compositions of salsalate
US8602215B2 (en) 2010-06-30 2013-12-10 Sanofi Methods for reducing the risk of an adverse dronedarone/beta-blockers interaction in a patient suffering from atrial fibrillation
WO2014065537A1 (en) * 2012-10-23 2014-05-01 씨지케이바이오 주식회사 Pharmaceutical composition for preventing and treating prostate cancer
WO2014068007A1 (en) * 2012-10-30 2014-05-08 Pharnext Compositions, methods and uses for the treatment of diabetes and related conditions by controlling blood glucose level
CN103880946A (en) * 2012-12-20 2014-06-25 深圳翰宇药业股份有限公司 Preparation method of Carperitide
US20150038513A1 (en) * 2011-07-04 2015-02-05 Les Laboratoires Servier Association between n-hydroxy-4-benzamide and folfox
US9427429B2 (en) 2010-03-01 2016-08-30 Tau Therapeutics Llc Cancer diagnosis and imaging
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9707184B2 (en) 2014-07-17 2017-07-18 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10617689B2 (en) 2013-10-30 2020-04-14 Pharnext Compositions, methods and uses for the treatment of diabetes and related conditions by controlling blood glucose level
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US11491142B2 (en) 2020-04-17 2022-11-08 Honeybrains, Llc Compositions and methods for treating neuropsychiatric disorders
CN116286669A (en) * 2023-03-23 2023-06-23 江南大学 Hybridoma cell strain secreting toraset Mi Shan clone antibody and application thereof
CN116622514A (en) * 2023-07-21 2023-08-22 南京师范大学 Regulation and control method for improving polyunsaturated fatty acid content in microbial thalli and/or microbial grease and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060199796A1 (en) * 2004-08-13 2006-09-07 Amgen Inc. Substituted benzofused heterocycles
US7115767B2 (en) * 2003-07-18 2006-10-03 Abbott Laboratories Tetraline derivatives as ghrelin receptor modulators
US20060293370A1 (en) * 2005-06-10 2006-12-28 Saunders Jeffrey O Sulfonamide compounds and uses thereof
US20070037857A1 (en) * 2005-08-15 2007-02-15 Zentaris Gmbh Novel triazole derivatives as ghrelin analogue ligands of growth hormone secretagogue receptors
US20070078135A1 (en) * 2005-04-18 2007-04-05 Neurogen Corporation Substituted heteroaryl CB1 antagonists

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7115767B2 (en) * 2003-07-18 2006-10-03 Abbott Laboratories Tetraline derivatives as ghrelin receptor modulators
US20060199796A1 (en) * 2004-08-13 2006-09-07 Amgen Inc. Substituted benzofused heterocycles
US20070078135A1 (en) * 2005-04-18 2007-04-05 Neurogen Corporation Substituted heteroaryl CB1 antagonists
US20060293370A1 (en) * 2005-06-10 2006-12-28 Saunders Jeffrey O Sulfonamide compounds and uses thereof
US20070037857A1 (en) * 2005-08-15 2007-02-15 Zentaris Gmbh Novel triazole derivatives as ghrelin analogue ligands of growth hormone secretagogue receptors

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9107900B2 (en) 2008-04-17 2015-08-18 Sanofi Use of dronedarone for the preparation of a medicament for use in the prevention of cardiovascular hospitalization or of morality
US8410167B2 (en) 2008-04-17 2013-04-02 Sanofi Use of dronedarone for the preparation of a medicament for use in the prevention of cardiovascular hospitalization or of mortality
JP2012528890A (en) * 2009-06-05 2012-11-15 ティーエーユー・セラピューティクス・エルエルシー Combination methods for treating cancer or precancerous conditions
JP2016028058A (en) * 2009-06-05 2016-02-25 ティーエーユー・セラピューティクス・エルエルシー Combination method for treating cancer or precancerous condition
WO2011008052A3 (en) * 2009-07-17 2011-05-19 한국생명공학연구원 Composition for the prevention or treatment of bone diseases comprising colforsin daropate
KR20110007978A (en) * 2009-07-17 2011-01-25 한국생명공학연구원 A composition for preventing and treating bone disease comprising colforsin daropate
KR101656834B1 (en) 2009-07-17 2016-09-13 한국생명공학연구원 A composition for preventing and treating bone disease comprising colforsin daropate
US8865762B2 (en) 2009-07-17 2014-10-21 Korea Research Institute Of Bioscience And Biotechnology Method for the treatment of bone diseases comprising colforsin daropate
US9427429B2 (en) 2010-03-01 2016-08-30 Tau Therapeutics Llc Cancer diagnosis and imaging
US8602215B2 (en) 2010-06-30 2013-12-10 Sanofi Methods for reducing the risk of an adverse dronedarone/beta-blockers interaction in a patient suffering from atrial fibrillation
US20150038513A1 (en) * 2011-07-04 2015-02-05 Les Laboratoires Servier Association between n-hydroxy-4-benzamide and folfox
WO2013175500A1 (en) * 2012-04-23 2013-11-28 Cadila Healthcare Limited Delazed release pharmaceutical compositions of salsalate
WO2014065537A1 (en) * 2012-10-23 2014-05-01 씨지케이바이오 주식회사 Pharmaceutical composition for preventing and treating prostate cancer
WO2014068007A1 (en) * 2012-10-30 2014-05-08 Pharnext Compositions, methods and uses for the treatment of diabetes and related conditions by controlling blood glucose level
EA031798B1 (en) * 2012-10-30 2019-02-28 Фарнекст Use of ifenprodil for the treatment of diabetes
US10596160B2 (en) 2012-10-30 2020-03-24 Pharnext Compositions, methods and uses for the treatment of diabetes and related conditions by controlling blood glucose level
US10092554B2 (en) 2012-10-30 2018-10-09 Pharnext Compositions, methods and uses for the treatment of diabetes and related conditions by controlling blood glucose level
EP3482753A1 (en) * 2012-10-30 2019-05-15 Pharnext Compositions, methods and uses for the treatment of diabetes and related conditions by controlling blood glucose level
CN103880946A (en) * 2012-12-20 2014-06-25 深圳翰宇药业股份有限公司 Preparation method of Carperitide
US10639281B2 (en) 2013-08-12 2020-05-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10617689B2 (en) 2013-10-30 2020-04-14 Pharnext Compositions, methods and uses for the treatment of diabetes and related conditions by controlling blood glucose level
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10792254B2 (en) 2013-12-17 2020-10-06 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9707184B2 (en) 2014-07-17 2017-07-18 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US11491142B2 (en) 2020-04-17 2022-11-08 Honeybrains, Llc Compositions and methods for treating neuropsychiatric disorders
US11583520B2 (en) 2020-04-17 2023-02-21 Hb Biotech, Inc. Compositions and methods for treating neuropsychiatric disorders
CN116286669A (en) * 2023-03-23 2023-06-23 江南大学 Hybridoma cell strain secreting toraset Mi Shan clone antibody and application thereof
CN116286669B (en) * 2023-03-23 2023-09-22 江南大学 Hybridoma cell strain secreting toraset Mi Shan clone antibody and application thereof
CN116622514A (en) * 2023-07-21 2023-08-22 南京师范大学 Regulation and control method for improving polyunsaturated fatty acid content in microbial thalli and/or microbial grease and application thereof
CN116622514B (en) * 2023-07-21 2023-10-20 南京师范大学 Regulation and control method for improving polyunsaturated fatty acid content in microbial thalli and/or microbial grease and application thereof

Similar Documents

Publication Publication Date Title
WO2008141189A1 (en) Ghrelin modulating compounds and combinations thereof
US7829589B2 (en) Sulfonamide compounds and uses thereof
US7897765B2 (en) Therapeutic compounds and uses thereof
US20050261332A1 (en) Sulfonamides and uses thereof
EP1893569A1 (en) Sulfonamide compounds and uses thereof
US20120088747A1 (en) Sulfonamide containing compounds and uses thereof
EP3782991A1 (en) Novel modulators of sphingosine phosphate receptors
JP2023513272A (en) heterocyclic GLP-1 agonists
US11046660B2 (en) Compounds and their use as PDE4 activators
US10385027B2 (en) Triazole derivatives and their use as PDE4 activators
EP1797090A2 (en) Sulfonamides and uses thereof
KR20190094167A (en) Treatment of CNS Disease with sGC Stimulants
CN108602775B (en) Mast cell modulators and uses thereof
KR20050106421A (en) Carboxylic acid compounds
JP2009513510A (en) Macrocyclic compound having aspartic protease inhibitory activity and pharmaceutical use thereof
CA3220005A1 (en) Modulators of g-protein coupled receptors
KR20180128930A (en) Benzenesulfonyl-asymmetric ureas and their medical uses
WO2010104929A1 (en) Sulfonamide containing compounds and uses thereof
WO2010104967A1 (en) Sulfonamide containing compounds and uses thereof

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: 08780627

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: 08780627

Country of ref document: EP

Kind code of ref document: A1