WO2018232158A1 - Méthodes de traitement de maladies métaboliques avec des pyrroles bicycliques fusionnés - Google Patents

Méthodes de traitement de maladies métaboliques avec des pyrroles bicycliques fusionnés Download PDF

Info

Publication number
WO2018232158A1
WO2018232158A1 PCT/US2018/037602 US2018037602W WO2018232158A1 WO 2018232158 A1 WO2018232158 A1 WO 2018232158A1 US 2018037602 W US2018037602 W US 2018037602W WO 2018232158 A1 WO2018232158 A1 WO 2018232158A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
formula
alkylene
compound
optionally substituted
Prior art date
Application number
PCT/US2018/037602
Other languages
English (en)
Inventor
Yu-Hsin Tom Lin
Cheng-Chi Irene Wang
Original Assignee
Belite Bio, 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 Belite Bio, Inc. filed Critical Belite Bio, Inc.
Publication of WO2018232158A1 publication Critical patent/WO2018232158A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • Some embodiments provided herein describe methods of treating a metabolic disease or disorder in a subject in need thereof, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a compound of Formula (II), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof:
  • is absent or present, and when present is a bond
  • each R 1 is independently halogen, haloalkyl, or alkyl
  • R 2 is absent or present, and when present is H, -OH, or halogen;
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • heterobicycle substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • the compound of Formula (II) has the structure:
  • B is a substituted or unsubstituted monocycle or substituted or unsubstituted heteromonocycle.
  • B is a substituted or unsubstituted imidazole, substituted or unsubstituted pyridazine, substituted or unsubstituted triazine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyridine, substituted or unsubstituted pyrimidine, substituted or unsubstituted pyrazine, substituted or unsubstituted thiadiazole, substituted or unsubstituted thiazole, substituted or unsubstituted pyrrole, substituted or unsubstituted oxazole, or substituted or unsubstituted triazole.
  • each R 1 is independently F, Br, Cl, C 1-6 haloalkyl, or C 1-6 alkyl.
  • each R 1 is independently F or CF 3 .
  • B has the structure:
  • each R 3 is independently H, F, Cl, halogen, -CF 3 , -OCH 3 , SO 2 CH 3 , -OH, NH 2 , C 1 -C 4 alkyl , C 3 -C 6 cycloalkyl, -O(C 1 -C 4 alkyl), -C(O)NH 2 , -C(O)NH(C 1 -C 10 alkyl), - C(O)N(C 1 -C 10 alkyl) 2 , -C(O)OH, -C(O)O(C 1 -C 4 alkyl), -C(O)(C 1 -C 4 alkyl), - C(O)NHSO 2 (C 1 -C 10 alkyl), -C(O)NH(C 3 -C 6 cycloalkyl), -C(O)NH(aryl), -OSO 2 NH 2 , -NHC(O)NH(C 1 -C 10 alkyl),
  • n 0, 1, 2, or 3.
  • a metabolic disease or disorder comprising a compound of Formula (II) wherein ⁇ is absent or is present; R 1 is independently selected from t-Bu, Cl, F, or–CF 3 ; and R 2 is absent or present, and when present is H, -OH, or F.
  • is absent; R 1 is–CF 3 ; and R 2 is H.
  • is present; R 1 is–CF 3 ; and R 2 is absent.
  • One embodiment provides a method of treating a metabolic disease or disorder in a subject in need thereof, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a com ound having the structure:
  • One embodiment provides a method of treating a metabolic disease or disorder in a subject in need thereof, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a compound of Formula (III), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof:
  • each R 1 is independently halogen, haloalkyl, or alkyl
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • substituted or unsubstituted heteromonocycle substituted or unsubstituted heterobicycle, substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • B has the structure:
  • ⁇ , ⁇ , ⁇ , and ⁇ are each independently absent or present, and when present each is a bond;
  • X is C
  • Z 1 is S, O, or N
  • Z 2 is S, O, N, or NR 3 ;
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • C is a substituted or unsubstituted fused 5-, 6-, or 7- membered ring structure.
  • a metabolic disease or disorder comprising a compound of Formula (III)
  • Z 1 when ⁇ is present, then Z 1 is O or S, Z 2 is N, X is C, ⁇ is present, and ⁇ and ⁇ are absent; when ⁇ is absent, then Z 1 is N, Z 2 is NR 3 , X is C, ⁇ and ⁇ are present, and ⁇ is absent; or when ⁇ is absent, then Z 1 is N, Z 2 is O or S, X is C, ⁇ and ⁇ are present, and ⁇ is absent.
  • B has the structure
  • n 0, 1, or 2;
  • ⁇ , ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ are each independently absent or present, and when present each is a bond;
  • Z 1 is S, O, or N
  • Z 2 is S, O, N or NR 3 ,
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • X is C
  • R 4 is H, halogen, C 1 -C 10 alkyl, C 1 -C 10 cycloalkyl, -O(C 1 -C 10 alkyl), -C(O)OH, - C(O)O(C 1 -C 10 alkyl), -C(O)NH 2 , -C(O)NH(C 1 -C 4 alkyl), -C(O)N(C 1 -C 4 alkyl) 2 , - NHC(O)NH(C 1 -C 10 alkyl), -NHC(O)N(C 1 -C 4 alkyl) 2 , -SO 2 NH(C 1 -C 10 alkyl), - SO 2 N(C 1 -C l0 alkyl) 2 , -CN, or -CF 3 ;
  • R 5 is H or C 1 -C 10 alkyl
  • R 6 is H, C 1 -C 10 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 10 alkylene)CF 3 , -(C 1 - C 10 alkylene)OCH 3 , -(C 1 -C 10 alkylene)-halogen, alkyl), -SO 2 (C 1 -C 10 alkylene) -CF 3 , -SO 2 (C 1 -C 10 alkylene)OCH 3 , -SO 2 (C 1 -C 10 alkylene)-halogen, - C(O)(C 1 -C 10 alkyl), -C(O)(C 1 -C 10 alkylene)CF 3 , -C(O)(C 1 -C 10 alkylene)OCH 3 , - C(O)(C 1 -C 10 alkylene)-halogen, -C(O)NH(C 1 -C 10 alkyl), -C(O)N(C 1
  • ⁇ and ⁇ are present; ⁇ , ⁇ , ⁇ , and ⁇ are absent; Z 1 is N; Z 2 is O, S, or NR 3 ; R 3 is H, C 1 -C 4 alkyl, or oxetane; and X is C.
  • B has the structure
  • n 0;
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • Y 1 and Y 3 are each CH 2 or C(CH 3 ) 2 ;
  • Y 2 is O, SO 2 , or NR 6 ;
  • R 6 is H, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 4 alkylene)CF 3 , -(C 1 -C 4 alkylene)OCH 3 , -(C 1 -C 4 alkylene)-halogen, -SO 2 (C 1 -C 4 alkyl), -SO 2 (C 1 -C 4 alkylene)CF 3 , -SO 2 (C 1 - C 4 alkylene)OCH 3 , -SO 2 (C 1 -C 4 alkylene)-halogen, -C(O)(C 1 -C 4 alkyl), -C(O)(C 1 - C 4 alkylene)CF 3 , -C(O)(C 1 -C 4 alkylene)OCH 3 , -C(O)(C 1 -C 4 alkylene)-halogen, - C(O)NH(C 1 -C 4 alkyl), -
  • n 1;
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • Y 1 and Y 4 are CH 2 or C(CH 3 ) 2 ;
  • Y 2 and Y 3 are each CH 2 or C(CH 3 ) 2 , O, SO 2 , or NR 6 ;
  • R 6 is H, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 4 alkylene)CF 3 , -(C 1 -C 4 alkylene)OCH 3 , -(C 1 -C 4 alkylene)-halogen, -SO 2 (C 1 -C 4 alkyl), -SO 2 (C 1 -C 4 alkylene)CF 3 , -SO 2 (C 1 -C 4 alkylene)OCH 3 , -SO 2 (C 1 -C 4 alkylene)-halogen, -C(O)(C 1 -C 4 alkyl), -C(O)(C 1 -C 4 alkylene)CF 3 , -C(O)(C 1 -C 4 alkylene)OCH 3 , -C(O)(C 1 -C 4 alkylene)-halogen, - C(O)NH(C 1 -C 4 alkyl), -
  • n 2;
  • R 3 is H, C 1 -C 4 alkyl, or oxetane; Y 1 and Y 4 are CH 2 or C(CH 3 ) 2 ;
  • Y 2 and Y 3 are each CH 2 or C(CH 3 ) 2 , O, SO 2 , or NR 6 ;
  • R 6 is H, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 4 alkylene)CF 3 , -(C 1 -C 4 alkylene)OCH 3 , -(C 1 -C 4 alkylene)-halogen, -SO 2 (C 1 -C 4 alkyl), -SO 2 (C 1 -C 4 alkylene)CF 3 , -SO 2 (C 1 -C 4 alkylene)OCH 3 , -SO 2 (C 1 -C 4 alkylene)-halogen, -C(O)(C 1 -C 4 alkyl), -C(O)(C 1 -C 4 alkylene)CF 3 , -C(O)(C 1 -C 4 alkylene)OCH 3 , -C(O)(C 1 -C 4 alkylene)-halogen, - C(O)NH(C 1 -C 4 alkyl), -
  • B has the structure:
  • B has the structure:
  • B has the structure:
  • R 6 is H, C 1 -C 4 alkyl, -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 CH(CH 3 ) 2 , t-Bu, - CH 2 OCH 3 , -CH 2 CF 3 , -CH 2 Cl, -CH 2 F, -CH 2 CH 2 OCH 3 , -CH 2 CH 2 CF 3 , -CH 2 CH 2 Cl, -CH 2 CH 2 F, or .
  • R 6 is -SO 2 CH 3 , -SO 2 CH 2 CH 3 , -SO 2 CH 2 CH 2 CH 3 , -SO 2 CH(CH 3 ) 2 , - SO 2 CH 2 CH(CH 3 ) 2 , -SO 2 (t-Bu), -SO 2 CH 2 OCH 3 , -SO 2 CH 2 CF 3 , -SO 2 CH 2 Cl, -SO 2 CH 2 F, -
  • R 6 is C(O)CH 3 , C(O)CH 2 CH 3 , -C(O)CH 2 CH 2 CH 3 , -C(O)CH(CH 3 ) 2 , - C(O)CH 2 CH(CH 3 ) 2 , -C(O)t-Bu, -C(O)CH 2 OCH 3 , -C(O)CH 2 CF 3 , -C(O)CH 2 Cl, -C(O)CH 2 F, -
  • B has the structure
  • ⁇ , ⁇ , ⁇ , and ⁇ are present; ⁇ , and ⁇ are absent; Z 1 is N; Z 2 is O or NR 3 ; R 3 is H, C 1 -C 4 alkyl, or oxetane; and X is C.
  • B has the structure
  • Y 1 , Y 2 , Y 3 and each occurrence of Y 4 are each independently CR 4 , or N;
  • R 3 is H, halogen, C 1 -C 10 alkyl, C 1 -C 10 cycloalkyl , -O(C 1 -C 10 alkyl), -C(O)OH, - C(O)O(C 1 -C 10 alkyl), -C(O)NH 2 , -C(O)NH(C 1 -C 4 alkyl), -C(O)N(C 1 -C 4 alkyl) 2 , - NHC(O)NH(C 1 -C 10 alkyl), -NHC(O)N(C 1 -C 4 alkyl) 2 , -SO 2 NH(C 1 -C 10 alkyl), - SO 2 N(C 1 -C l0 alkyl) 2 , -CN, or -CF 3 .
  • Y 1 , Y 2 , Y 3 and Y 4 are CH;Y 1 , Y 2 , Y 3 are CH and Y 4 is N;Y 1 , Y 2 , Y 4 are CH and Y 3 is N;Y 1 , Y 3 , Y 4 are CH and Y 2 is N; or Y 2 , Y 3 , Y 4 are CH and Y 1 is N.
  • R 3 is H, halogen, C 1 -C 10 alkyl, C 1 -C 10 cycloalkyl , -O(C 1 -C 10 alkyl), -C(O)OH, - C(O)O(C 1 -C 10 alkyl), -C(O)NH 2 , -C(O)NH(C 1 -C 4 alkyl), -C(O)N(C 1 -C 4 alkyl) 2 , - NHC(O)NH(C 1 -C 10 alkyl), -NHC(O)N(C 1 -C 4 alkyl) 2 , -SO 2 NH(C 1 -C 10 alkyl), - SO 2 N(C 1 -C l0 alkyl) 2 , -CN, or -CF 3 ; and
  • R 4 is H, halogen, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, -O(C 1 -C 4 alkyl), -CN, -CF 3 , - C(O)OH, -C(O)NH 2 , -C(O)N(CH 3 ) 2 , -C(O)NHCH 3 , or -NHC(O)N(CH 3 ) 2 .
  • each R 4 is independently F, Br, Cl, C 1-6 haloalkyl, or alkyl.
  • each R 1 is independently F, Br, Cl, C 1-6 haloalkyl, or C 1-6 alkyl.
  • One embodiment provides a method of treating a metabolic disease or disorder in a subject in need thereof, the methods comprising administering to the subject a composition comprising a therapeutically effective amount of a compound having the structure: , or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof.
  • the metabolic disease or disorder is obesity, type II diabetes, diabetic retinopathy, non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), liver fibrosis, liver cancer, or liver cirrhosis.
  • the therapeutically effective amount of a compound of Formula (II) or Formula (III), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is about 25 mg per day, about 50 mg per day, about 75 mg per day, about 100 mg per day, about 150 mg per day, about 200 mg per day, or about 400 mg per day.
  • the therapeutically effective amount of a compound of Formula (II) or Formula (III), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is about 50 mg per day, about 100 mg per day, about 150 mg per day, about 200 mg per day, or about 400 mg per day.
  • the therapeutically effective amount of a compound of Formula (II) or Formula (III), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is up to 25 mg per day, up to 50 mg per day, up to 75 mg per day, up to 100 mg per day, up to 150 mg per day, up to 200 mg per day, up to 400 mg per day, up to 600 mg per day, up to 800 mg per day, or up to 1000 mg per day.
  • the therapeutically effective amount of a compound of Formula (II) or Formula (III), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is up to 400 mg per day.
  • the serum or plasma levels of RBP4 are reduced by at least 50% from baseline. In some embodiments, 24 hours after administration of a compound of Formula (II) or Formula (III), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 65% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 80% from baseline. In some embodiments, 24 hours after administration of a compound of Formula (II) or Formula (III), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 85% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 1 mg/dL. In some embodiments, 24 hours after administration of a compound of Formula (II) or Formula (III), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 2 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 5 mg/dL. In some embodiments, 24 hours after administration of a compound of Formula (II) or Formula (III), a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 10 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 15 mg/dL.
  • the metabolic disease or disorder is obesity. In some embodiments, the metabolic disease or disorder is type II diabetes. In some embodiments, the metabolic disease or disorder is non-proliferative diabetic retinopathy (NPDR) or proliferative diabetic retinopathy (PDR). In some embodiments, the metabolic disease or disorder is a liver disease. In some embodiments, the metabolic disease or disorder is non-alcoholic fatty liver disease (NAFLD). In some embodiments, the metabolic disease or disorder is non-alcoholic steatohepatitis (NASH). In some embodiments, the metabolic disease or disorder is liver fibrosis. In some embodiments, the metabolic disease or disorder is liver cirrhosis. In some embodiments, the metabolic disease or disorder is liver cancer.
  • NPDR non-proliferative diabetic retinopathy
  • PDR proliferative diabetic retinopathy
  • the metabolic disease or disorder is a liver disease. In some embodiments, the metabolic disease or disorder is non-alcoholic fatty liver disease (NAFLD). In some embodiments, the metabolic disease
  • the compound of Formula (II) or Formula (III) is administered to the subject orally or intravenously. In certain embodiments, the compound of Formula (II) or Formula (III) is administered to the subject orally. In certain embodiments, the compound of Formula (II) or Formula (III) is administered to the subject intravenously. In some embodiments, administration of a compound of Formula (II) or Formula (III) to a subject in need thereof is to treat an existing disease or disorder. In some embodiments, the administration of a compound of Formula (II) or Formula (III) is provided as prophylaxis. INCORPORATION BY REFERENCE
  • Fig.1 illustrates absolute values of RBP4 plasma levels in non-human primates following a single intravenous dose of vehicle control.
  • Fig.2 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in non-human primates following a single oral dose of vehicle control.
  • Fig.3 illustrates absolute values of RBP4 plasma levels in non-human primates following a single intravenous dose of Compound 1 at 1 mg/kg.
  • Fig.4 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in non-human primates following a single intravenous dose of Compound 1 at 1 mg/kg.
  • Fig.5 illustrates absolute values of RBP4 plasma levels in non-human primates following a single oral dose of Compound 1 at 5 mg/kg.
  • Fig.6 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in non-human primates following a single oral dose of Compound 1 at 5 mg/kg.
  • Fig.7 illustrates Compound 1 plasma levels in non-human primates following a single intravenous dose of 1 mg/kg.
  • Fig.8 illustrates Compound 1 plasma levels in non-human primates following a single oral dose of 5 mg/kg.
  • Fig.9 illustrates absolute values of RBP4 plasma levels in male Sprague Dawley rats following a single intravenous dose of Compound 1 at 2 mg/kg.
  • Fig.10 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in male Sprague Dawley rats following a single intravenous dose of Compound 1 at 2 mg/kg.
  • Fig.11 illustrates absolute values of RBP4 plasma levels in male Sprague Dawley rats following a single oral dose of Compound 1 at 5 mg/kg.
  • Fig.12 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in male Sprague Dawley rats following a single oral dose of Compound 1 at 5 mg/kg.
  • Fig.13 illustrates Compound 1 plasma levels in male Sprague Dawley rats following a single intravenous dose of 2 mg/kg.
  • Fig.14 illustrates Compound 1 plasma levels in male Sprague Dawley rats following a single oral dose of 5 mg/kg.
  • Fig.15 illustrates absolute values of RBP4 plasma levels in male Sprague Dawley rats following a single oral dose of Compound 1 at 5 mg/kg on day 1 of treatment.
  • Fig.16 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in male Sprague Dawley rats following a single oral dose of Compound 1 at 5 mg/kg on day 1 of treatment.
  • Fig.17 illustrates absolute values of RBP4 plasma levels in male Sprague Dawley rats following a single oral dose of vehicle control on day 1 of treatment.
  • Fig.18 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in male Sprague Dawley rats following a single oral dose vehicle control on day 1 of treatment.
  • Fig.19 illustrates Compound 1 plasma levels in male Sprague Dawley rats following a single oral dose of 5 mg/kg on day 1 of treatment.
  • Fig.20 illustrates absolute values of pre-dose RBP4 plasma levels during a 7 day oral Compound 1 administration in male Sprague Dawley rats at 5 mg/kg.
  • Fig.21 illustrates percent reduction of pre-dose RBP4 plasma levels compared to day 1 pre- dose levels during a 7 day oral Compound 1 administration in male Sprague Dawley rats at 5 mg/kg.
  • Fig.22 illustrates Compound 1 plasma levels at pre-dose timepoints on days 1-7 of a 7 day oral Compound 1 administration in male Sprague Dawley rats at 5 mg/kg.
  • Fig.23 illustrates RBP4 pre-dose plasma levels during a 7 day oral vehicle administration in male Sprague Dawley rats.
  • Fig.24 illustrates absolute values of pre-dose RBP4 plasma levels on day 7 following a 7 day oral Compound 1 administration in male Sprague Dawley rats at 5 mg/kg.
  • Fig.25 illustrates percent reduction of pre-dose RBP4 plasma levels on day 7 compared to day 1 pre-dose levels during a 7 day oral Compound 1 administration in male Sprague Dawley rats at 5 mg/kg.
  • Fig.26 illustrates Compound 1 plasma levels on day 7 following a 7 day oral Compound 1 administration in male Sprague Dawley rats at 5 mg/kg.
  • Fig.27 illustrates absolute values of RBP4 plasma levels in male mice following a single intravenous dose of Compound 1 at 2 mg/kg.
  • Fig.28 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in male mice following a single intravenous dose of Compound 1 at 2 mg/kg.
  • Fig.29 illustrates absolute values of RBP4 plasma levels in male mice following a single oral dose of Compound 1 at 5 mg/kg.
  • Fig.30 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in male mice following a single oral dose of Compound 1 at 5 mg/kg.
  • Fig.31 illustrates Compound 1 plasma levels in male mice following a single intravenous dose of 2 mg/kg.
  • Fig.32 illustrates Compound 1 plasma levels in male mice following a single oral dose of 5 mg/kg.
  • Fig.33 illustrates absolute values of RBP4 plasma levels in male beagle dogs following a single intravenous dose of Compound 1 at 0.5 mg/kg.
  • Fig.34 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in male beagle dogs following a single intravenous dose of Compound 1 at 0.5 mg/kg.
  • Fig.35 illustrates absolute values of RBP4 serum levels in male beagle dogs following a single oral dose of Compound 1 at 2 mg/kg.
  • Fig.36 illustrates the percent reduction of RBP4 plasma levels compared to pre-dose levels in male beagle dogs following a single oral dose of Compound 1 at 2 mg/kg.
  • Fig.37 illustrates Compound 1 plasma levels in male beagle dogs following a single intravenous dose of 0.5 mg/kg.
  • Fig.38 illustrates Compound 1 plasma levels in male beagle dogs following a single oral dose of 2 mg/kg.
  • Niro refers to the -NO 2 radical.
  • Oxa refers to the -O- radical.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to fifteen carbon atoms (e.g., C 1 - C 15 alkyl).
  • an alkyl comprises one to thirteen carbon atoms (e.g., C 1 -C 13 alkyl).
  • an alkyl comprises one to eight carbon atoms (e.g., C 1 -C 8 alkyl).
  • an alkyl comprises one to five carbon atoms (e.g., C 1 -C 5 alkyl).
  • an alkyl comprises one to four carbon atoms (e.g., C 1 -C 4 alkyl). In other embodiments, an alkyl comprises one to three carbon atoms (e.g., C 1 -C 3 alkyl). In other embodiments, an alkyl comprises one to two carbon atoms (e.g., C 1 -C 2 alkyl). In other embodiments, an alkyl comprises one carbon atom (e.g., C 1 alkyl). In other embodiments, an alkyl comprises five to fifteen carbon atoms (e.g., C 5 -C 15 alkyl). In other embodiments, an alkyl comprises five to eight carbon atoms (e.g., C 5 -C 8 alkyl).
  • an alkyl comprises two to five carbon atoms (e.g., C 2 -C 5 alkyl). In other embodiments, an alkyl comprises three to five carbon atoms (e.g., C 3 -C 5 alkyl). In other embodiments, the alkyl group is selected from methyl, ethyl, 1-propyl (n-propyl), 1-methylethyl (iso-propyl), 1-butyl (n-butyl), 1-methylpropyl (sec-butyl), 2-methylpropyl (iso-butyl),
  • alkyl is attached to the rest of the molecule by a single bond.
  • an alkyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , -SR a , -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -C(O)N(R a ) 2 , - N(R a )C(O)OR a , -OC(O)-N(R a ) 2 , -N(R a )C(O)R a , -N(R a )
  • Alkoxy refers to a radical bonded through an oxygen atom of the formula–O-alkyl, where alkyl is an alkyl chain as defined above.
  • alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one carbon-carbon double bond, and having from two to twelve carbon atoms. In certain embodiments, an alkenyl comprises two to eight carbon atoms. In other embodiments, an alkenyl comprises two to four carbon atoms. The alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl), prop-1-enyl (i.e., allyl), but-1-enyl, pent-1-enyl, penta-1,4-dienyl, and the like.
  • an alkenyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , -SR a , - OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -C(O)N(R a ) 2 , -N(R a )C(O)OR a , -OC(O)-N(R a ) 2 , - N(R a )C(O)R a , -N(R a )S(O) t R a (where t is 1 or 2), -S(O) t OR a (where t is 1 or 2), -S(O) t R
  • Alkynyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one carbon-carbon triple bond, having from two to twelve carbon atoms.
  • an alkynyl comprises two to eight carbon atoms.
  • an alkynyl comprises two to six carbon atoms.
  • an alkynyl comprises two to four carbon atoms.
  • the alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
  • an alkynyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , - SR a , -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -C(O)N(R a ) 2 , -N(R a )C(O)OR a , -OC(O)-N(R a ) 2 , - N(R a )C(O)R a , -N(R a )S(O) t R a (where t is 1 or 2), -S(O) t OR a (where t is 1 or 2), -S(O) t
  • Alkylene or "alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms, for example, methylene, ethylene, propylene, n-butylene, and the like.
  • the alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
  • the points of attachment of the alkylene chain to the rest of the molecule and to the radical group are through one carbon in the alkylene chain or through any two carbons within the chain.
  • an alkylene comprises one to eight carbon atoms (e.g., C 1 -C 8 alkylene). In other embodiments, an alkylene comprises one to five carbon atoms (e.g., C 1 -C 5 alkylene). In other embodiments, an alkylene comprises one to four carbon atoms (e.g., C 1 -C 4 alkylene). In other embodiments, an alkylene comprises one to three carbon atoms (e.g., C 1 -C 3 alkylene). In other embodiments, an alkylene comprises one to two carbon atoms (e.g., C 1 -C 2 alkylene). In other embodiments, an alkylene comprises one carbon atom (e.g., C 1 alkylene).
  • an alkylene comprises five to eight carbon atoms (e.g., C 5 -C 8 alkylene). In other embodiments, an alkylene comprises two to five carbon atoms (e.g., C 2 -C 5 alkylene). In other embodiments, an alkylene comprises three to five carbon atoms (e.g., C 3 -C 5 alkylene).
  • an alkylene chain is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , -SR a , -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , -C(O)N(R a ) 2 , -N(R a )C(O)OR a , -OC(O)- N(R a ) 2 , -N(R a )C(O)R a , - N(R a )S(O) t R a (where t is 1 or 2), -S(O) t OR a (where t is 1 or 2), -S(O) t R a
  • alkenylene or "alkenylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon double bond, and having from two to twelve carbon atoms.
  • the alkenylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
  • an alkenylene comprises two to eight carbon atoms (e.g., C 2 -C 8 alkenylene).
  • an alkenylene comprises two to five carbon atoms (e.g., C 2 -C 5 alkenylene).
  • an alkenylene comprises two to four carbon atoms (e.g., C 2 -C 4 alkenylene). In other embodiments, an alkenylene comprises two to three carbon atoms (e.g., C 2 -C 3 alkenylene). In other embodiments, an alkenylene comprises five to eight carbon atoms (e.g., C 5 -C 8 alkenylene). In other embodiments, an alkenylene comprises two to five carbon atoms (e.g., C 2 -C 5 alkenylene). In other embodiments, an alkenylene comprises three to five carbon atoms (e.g., C 3 -C 5 alkenylene).
  • an alkenylene chain is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , -SR a , -OC(O)-R a , -N(R a ) 2 , -C(O)R a , - C(O)OR a , -C(O)N(R a ) 2 , -N(R a )C(O)OR a , -OC(O)- N(R a ) 2 , -N(R a )C(O)R a , -N(R a )S(O) t R a (where t is 1 or 2), -S(O) t OR a (where t is 1 or 2), -S(O) t R a (where t is 1 or
  • heterocyclylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl), heteroaryl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl), or heteroarylalkyl (optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl).
  • Alkynylene or “alkynylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing at least one carbon-carbon triple bond, and having from two to twelve carbon atoms.
  • the alkynylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
  • an alkynylene comprises two to eight carbon atoms (e.g., C 2 -C 8 alkynylene).
  • an alkynylene comprises two to five carbon atoms (e.g., C 2 -C 5 alkynylene).
  • an alkynylene comprises two to four carbon atoms (e.g., C 2 -C 4 alkynylene). In other embodiments, an alkynylene comprises two to three carbon atoms (e.g., C 2 -C 3 alkynylene). In other embodiments, an alkynylene comprises two carbon atom (e.g., C 2 alkylene). In other embodiments, an alkynylene comprises five to eight carbon atoms (e.g., C 5 -C 8 alkynylene). In other embodiments, an alkynylene comprises three to five carbon atoms (e.g., C 3 -C 5 alkynylene).
  • an alkynylene chain is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , -SR a , -OC(O)-R a , -N(R a ) 2 , -C(O)R a , -C(O)OR a , - C(O)N(R a ) 2 , -N(R a )C(O)OR a , -OC(O)-N(R a ) 2 , -N(R a )C(O)R a , -N(R a )S(O) t R a (where t is 1 or 2), - S(O) t OR a (where t is 1 or 2), -S(O) t R
  • Aryl refers to a radical derived from an aromatic monocyclic or multicyclic hydrocarbon ring system by removing a hydrogen atom from a ring carbon atom.
  • the aromatic monocyclic or multicyclic hydrocarbon ring system contains only hydrogen and carbon from five to eighteen carbon atoms, where at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) ⁇ –electron system in accordance with the Hückel theory.
  • the ring system from which aryl groups are derived include, but are not limited to, groups such as benzene, fluorene, indane, indene, tetralin and naphthalene.
  • aryl or the prefix “ar-” (such as in “aralkyl”) is meant to include aryl radicals optionally substituted by one or more substituents independently selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted
  • each R b is independently a direct bond or a straight or branched alkylene or alkenylene chain
  • R c is a straight or branched alkylene or alkenylene chain
  • Aralkyl refers to a radical of the formula -R c -aryl where R c is an alkylene chain as defined above, for example, methylene, ethylene, and the like.
  • the alkylene chain part of the aralkyl radical is optionally substituted as described above for an alkylene chain.
  • the aryl part of the aralkyl radical is optionally substituted as described above for an aryl group.
  • alkenyl refers to a radical of the formula–R d -aryl where R d is an alkenylene chain as defined above.
  • the aryl part of the aralkenyl radical is optionally substituted as described above for an aryl group.
  • the alkenylene chain part of the aralkenyl radical is optionally substituted as defined above for an alkenylene group.
  • Aralkynyl refers to a radical of the formula -R e -aryl, where R e is an alkynylene chain as defined above.
  • the aryl part of the aralkynyl radical is optionally substituted as described above for an aryl group.
  • the alkynylene chain part of the aralkynyl radical is optionally substituted as defined above for an alkynylene chain.
  • Alkoxy refers to a radical bonded through an oxygen atom of the formula - O-R c -aryl where R c is an alkylene chain as defined above, for example, methylene, ethylene, and the like.
  • the alkylene chain part of the aralkyl radical is optionally substituted as described above for an alkylene chain.
  • the aryl part of the aralkyl radical is optionally substituted as described above for an aryl group.
  • Carbocyclyl refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which includes fused or bridged ring systems, having from three to fifteen carbon atoms.
  • a carbocyclyl comprises three to ten carbon atoms.
  • a carbocyclyl comprises five to seven carbon atoms.
  • the carbocyclyl is attached to the rest of the molecule by a single bond.
  • Carbocyclyl is saturated (i.e., containing single C-C bonds only) or unsaturated (i.e., containing one or more double bonds or triple bonds).
  • a fully saturated carbocyclyl radical is also referred to as
  • cycloalkyl examples include, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • An unsaturated carbocyclyl is also referred to as “cycloalkenyl.”
  • monocyclic cycloalkenyls include, e.g., cyclopentenyl,
  • Polycyclic carbocyclyl radicals include, for example, adamantyl, norbornyl (i.e., bicyclo[2.2.1]heptanyl), norbornenyl, decalinyl,
  • carbocyclyl is meant to include carbocyclyl radicals that are optionally substituted by one or more substituents independently selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -R b -OR a , - R b -OC(O)-R a , -R b -OC(
  • heterocyclyl optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl
  • heterocyclylalkyl optionally substituted with halogen, hydroxy, methoxy, or trifluoromethyl
  • heteroaryl optionally substituted with halogen, hydroxy, methoxy, or
  • each R b is independently a direct bond or a straight or branched alkylene or alkenylene chain
  • R c is a straight or branched alkylene or alkenylene chain, and where each of the above substituents is unsubstituted unless otherwise indicated.
  • Carbocyclylalkyl refers to a radical of the formula–R c -carbocyclyl where R c is an alkylene chain as defined above.
  • R c is an alkylene chain as defined above.
  • the alkylene chain and the carbocyclyl radical are optionally substituted as defined above.
  • Carbocyclylalkynyl refers to a radical of the formula–R c -carbocyclyl where R c is an alkynylene chain as defined above.
  • R c is an alkynylene chain as defined above.
  • the alkynylene chain and the carbocyclyl radical are optionally substituted as defined above.
  • Carbocyclylalkoxy refers to a radical bonded through an oxygen atom of the formula–O-R c -carbocyclyl where R c is an alkylene chain as defined above.
  • R c is an alkylene chain as defined above.
  • the alkylene chain and the carbocyclyl radical are optionally substituted as defined above.
  • carboxylic acid bioisostere refers to a functional group or moiety that exhibits similar physical, biological and/or chemical properties as a carboxylic acid moiety.
  • carboxylic acid bioisosteres include, but are not limited to,
  • Halo or "halogen” refers to bromo, chloro, fluoro or iodo substituents.
  • Fluoroalkyl refers to an alkyl radical, as defined above, that is substituted by one or more fluoro radicals, as defined above, for example, trifluoromethyl, difluoromethyl, fluoromethyl, 2,2,2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, and the like.
  • the alkyl part of the fluoroalkyl radical is optionally substituted as defined above for an alkyl group.
  • Heterocyclyl refers to a stable 3- to 18-membered non-aromatic ring radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which optionally includes fused or bridged ring systems. The heteroatoms in the heterocyclyl radical are optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heterocyclyl radical is partially or fully saturated. The heterocyclyl is attached to the rest of the molecule through any atom of the ring(s).
  • heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl,
  • 2-oxopiperidinyl 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl,
  • heterocyclyl is meant to include heterocyclyl radicals as defined above that are optionally substituted by one or more substituents selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -R b -OR a , -R b -OC(O)-R a , - R b -OC(O)-OR a , -R b -OC(O)-N(R a ) 2 , -R
  • each R b is independently a direct bond or a straight or branched alkylene or alkenylene chain
  • R c is a straight or branched alkylene or alkenylene chain
  • N-heterocyclyl or“N-attached heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical.
  • An N-heterocyclyl radical is optionally substituted as described above for heterocyclyl radicals.
  • N-heterocyclyl radicals include, but are not limited to, 1-morpholinyl, 1- piperidinyl, 1-piperazinyl, 1-pyrrolidinyl, pyrazolidinyl, imidazolinyl, and imidazolidinyl.
  • C-heterocyclyl or“C-attached heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one heteroatom and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a carbon atom in the heterocyclyl radical.
  • a C-heterocyclyl radical is optionally substituted as described above for heterocyclyl radicals. Examples of such C-heterocyclyl radicals include, but are not limited to, 2-morpholinyl, 2- or 3- or 4-piperidinyl, 2-piperazinyl, 2- or 3-pyrrolidinyl, and the like.
  • Heterocyclylalkyl refers to a radical of the formula–R c -heterocyclyl where R c is an alkylene chain as defined above. If the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom.
  • the alkylene chain of the heterocyclylalkyl radical is optionally substituted as defined above for an alkylene chain.
  • the heterocyclyl part of the heterocyclylalkyl radical is optionally substituted as defined above for a heterocyclyl group.
  • Heterocyclylalkoxy refers to a radical bonded through an oxygen atom of the formula–O-R c -heterocyclyl where R c is an alkylene chain as defined above. If the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom.
  • the alkylene chain of the heterocyclylalkoxy radical is optionally substituted as defined above for an alkylene chain.
  • the heterocyclyl part of the heterocyclylalkoxy radical is optionally substituted as defined above for a heterocyclyl group.
  • Heteroaryl refers to a radical derived from a 3- to 18-membered aromatic ring radical that comprises two to seventeen carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur.
  • the heteroaryl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, wherein at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) ⁇ –electron system in accordance with the Hückel theory.
  • Heteroaryl includes fused or bridged ring systems.
  • the heteroatom(s) in the heteroaryl radical is optionally oxidized.
  • heteroaryl is attached to the rest of the molecule through any atom of the ring(s).
  • heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1,3-benzodioxolyl, benzofuranyl, benzooxazolyl, benzo[d]thiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, benzo[b][1,4]oxazinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothienyl (benzothion
  • pyrido[3,4-d]pyrimidinyl pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, 5,6,7,8-tetrahydroquinazolinyl,
  • heteroaryl is meant to include heteroaryl radicals as defined above which are optionally substituted by one or more substituents selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, haloalkenyl, haloalkynyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -R b -OR a , -R b -OC(O)-R a , - R b -OC(O)-R a , - R b -OC(O)-R
  • each R b is independently a direct bond or a straight or branched alkylene or alkenylene chain
  • R c is a straight or branched alkylene or alkenylene chain
  • N-heteroaryl refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical.
  • An N-heteroaryl radical is optionally substituted as described above for heteroaryl radicals.
  • C-heteroaryl refers to a heteroaryl radical as defined above and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a carbon atom in the heteroaryl radical.
  • a C-heteroaryl radical is optionally substituted as described above for heteroaryl radicals.
  • Heteroarylalkyl refers to a radical of the formula–R c -heteroaryl, where R c is an alkylene chain as defined above. If the heteroaryl is a nitrogen-containing heteroaryl, the heteroaryl is optionally attached to the alkyl radical at the nitrogen atom.
  • heteroarylalkyl radical is optionally substituted as defined above for an alkylene chain.
  • the heteroaryl part of the heteroarylalkyl radical is optionally substituted as defined above for a heteroaryl group.
  • Heteroarylalkoxy refers to a radical bonded through an oxygen atom of the formula –O-R c -heteroaryl, where R c is an alkylene chain as defined above. If the heteroaryl is a
  • the heteroaryl is optionally attached to the alkyl radical at the nitrogen atom.
  • the alkylene chain of the heteroarylalkoxy radical is optionally substituted as defined above for an alkylene chain.
  • the heteroaryl part of the heteroarylalkoxy radical is optionally substituted as defined above for a heteroaryl group.
  • the compounds disclosed herein in some embodiments, contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that are defined, in terms of absolute stereochemistry, as (R)- or (S)-. Unless stated otherwise, it is intended that all stereoisomeric forms of the compounds disclosed herein are contemplated by this disclosure. When the compounds described herein contain alkene double bonds, and unless specified otherwise, it is intended that this disclosure includes both E and Z geometric isomers (e.g., cis or trans.) Likewise, all possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included.
  • geometric isomer refers to E or Z geometric isomers (e.g., cis or trans) of an alkene double bond.
  • positional isomer refers to structural isomers around a central ring, such as ortho-, meta-, and para- isomers around a benzene ring.
  • a "tautomer” refers to a molecule wherein a proton shift from one atom of a molecule to another atom of the same molecule is possible.
  • the compounds disclosed herein are used in different enriched isotopic forms, e.g., enriched in the content of 2 H, 3 H, 11 C, 13 C and/or 14 C.
  • the compound is deuterated in at least one position.
  • deuterated forms can be made by the procedure described in U.S. Patent Nos.5,846,514 and 6,334,997.
  • deuteration can improve the metabolic stability and or efficacy, thus increasing the duration of action of drugs.
  • structures depicted herein are intended to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13 C- or 14 C-enriched carbon are within the scope of the present disclosure.
  • the compounds of the present disclosure optionally contain unnatural proportions of atomic isotopes at one or more atoms that constitute such compounds.
  • the compounds may be labeled with isotopes, such as for example, deuterium ( 2 H), tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C).
  • isotopes such as for example, deuterium ( 2 H), tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C).
  • Isotopic substitution with 2 H, 11 C, 13 C, 14 C, 15 C, 12 N, 13 N, 15 N, 16 N, 16 O, 17 O, 14 F, 15 F, 16 F, 17 F, 18 F, 33 S, 34 S, 35 S, 36 S, 35 Cl, 37 Cl, 79 Br, 81 Br, 125 I are all contemplated. All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention.
  • the compounds disclosed herein have some or all of the 1 H atoms replaced with 2 H atoms.
  • the methods of synthesis for deuterium-containing compounds are known in the art and include, by way of non-limiting example only, the following synthetic methods.
  • Deuterium substituted compounds are synthesized using various methods such as described in: Dean, Dennis C.; Editor. Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development. [In: Curr., Pharm. Des., 2000; 6(10)] 2000, 110 pp; George W.; Varma, Rajender S. The Synthesis of Radiolabeled Compounds via Organometallic Intermediates, Tetrahedron, 1989, 45(21), 6601-21; and Evans, E. Anthony. Synthesis of radiolabeled compounds, J. Radioanal. Chem., 1981, 64(1-2), 9-32.
  • Deuterated starting materials are readily available and are subjected to the synthetic methods described herein to provide for the synthesis of deuterium-containing compounds. Large numbers of deuterium-containing reagents and building blocks are available commercially from chemical vendors, such as Aldrich Chemical Co.
  • CD 3 I iodomethane-d 3
  • Deuterium-transfer reagents such as lithium aluminum deuteride (LiAlD 4 ) are employed to transfer deuterium under reducing conditions to the reaction substrate.
  • LiAlD 4 lithium aluminum deuteride
  • Deuterium gas and palladium catalyst are employed to reduce unsaturated carbon- carbon linkages and to perform a reductive substitution of aryl carbon-halogen bonds as illustrated, by way of example only, in the reaction schemes below.
  • the compounds disclosed herein contain one deuterium atom. In another embodiment, the compounds disclosed herein contain two deuterium atoms. In another embodiment, the compounds disclosed herein contain three deuterium atoms. In another
  • the compounds disclosed herein contain four deuterium atoms. In another embodiment, the compounds disclosed herein contain four deuterium atoms. In another
  • the compounds disclosed herein contain five deuterium atoms. In another embodiment, the compounds disclosed herein contain six deuterium atoms. In another embodiment, the compounds disclosed herein contain more than six deuterium atoms. In another embodiment, the compound disclosed herein is fully substituted with deuterium atoms and contains no non- exchangeable 1 H hydrogen atoms. In one embodiment, the level of deuterium incorporation is determined by synthetic methods in which a deuterated synthetic building block is used as a starting material.
  • “Pharmaceutically acceptable salt” includes both acid and base addition salts.
  • a pharmaceutically acceptable salt of any one of the heterocyclic RBP4 inhibitory compounds described herein is intended to encompass any and all pharmaceutically suitable salt forms.
  • Preferred pharmaceutically acceptable salts of the compounds described herein are pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, hydroiodic acid, hydrofluoric acid, phosphorous acid, and the like. Also included are salts that are formed with organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and. aromatic sulfonic acids, etc.
  • acetic acid trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
  • Exemplary salts thus include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, trifluoroacetates, propionates, caprylates, isobutyrates, oxalates, malonates, succinate suberates, sebacates, fumarates, maleates, mandelates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, phthalates, benzenesulfonates, toluenesulfonates, phenylacetates, citrates, lactates, malates, tartrates, methanesulfonates, and the like.
  • Acid addition salts of basic compounds are, in some embodiments, prepared by contacting the free base forms with a sufficient amount of the desired acid to produce the salt according to methods and techniques with which a skilled artisan is familiar.
  • “Pharmaceutically acceptable base addition salt” refers to those salts that retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Pharmaceutically acceptable base addition salts are, in some embodiments, formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, for example, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine,
  • treatment or“treating,” or“palliating” or“ameliorating” are used interchangeably. These terms refer to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the
  • compositions are, in some embodiments, administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease has not been made.
  • Prodrug is meant to indicate a compound that is, in some embodiments, converted under physiological conditions or by solvolysis to a biologically active compound described herein.
  • prodrug refers to a precursor of a biologically active compound that is
  • a prodrug is typically inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp.7-9, 21-24 (Elsevier, Amsterdam).
  • prodrugs are provided in Higuchi, T., et al., "Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol.14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
  • prodrug is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of an active compound, as described herein are prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound.
  • Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol or amine functional groups in the active compounds and the like.
  • RBP4 inhibitory compounds are provided herein in some embodiments.
  • the subject compounds and compositions comprising said compounds.
  • compositions are useful for inhibiting RPB4 and for the treatment of metabolic diseases or disorders, such as NASH, NAFLD, type II diabetes, obesity, fibrosis, cirrhosis, or hepatocellular carcinoma.
  • metabolic diseases or disorders such as NASH, NAFLD, type II diabetes, obesity, fibrosis, cirrhosis, or hepatocellular carcinoma.
  • Some embodiments provided herein describe a compound, or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, for use in treating a metabolic disease or disorder, having the structure of Formula (I):
  • U is C, CR 2 , or N;
  • is absent or present, and when present is a bond
  • each R 1 is independently halogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-CN, optionally substituted (C 0 -C 4 alkylene)-OR 7 , optionally substituted (C 0 -C 4 alkylene)-N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 8 )-COR 7 , optionally substituted (C 0 -C 4 alkylene)- SO 2 N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-SO 2 R 7 , optionally substituted (C 0 - C 4 alkylene)N(R 8 )-SO 2 N(R 7 ) 2 , or optionally substituted (C 0
  • each R 7 is independently selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, or optionally substituted heteroarylalkyl; or two R 11 groups together with the nitrogen to which they are attached join to form an optionally substituted N-heterocyclyl;
  • each R 8 is independently selected from H or optionally substituted alkyl;
  • R 2 is absent or present, and when present is -H, -OH, optionally substituted alkyl, or halogen;
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • heterobicycle substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • U is C and ⁇ is a bond. In other embodiments, U is CR 2 and ⁇ is absent. In some embodiments, U is N and ⁇ is absent.
  • Some embodiments provided herein describe a compound, or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, having the structure of Formula (I) wherein:
  • U is C, CR 2 , or N;
  • each R 1 is independently halogen, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 2-6 heterocyclyl, optionally substituted C 3-10 heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-CN, optionally substituted (C 0 -C 4 alkylene)-OR 7 , optionally substituted (C 0 -C 4 alkylene)- N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 8 )-COR 7 , optionally substituted (C 0 -C 4 alkylene)-SO 2 N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-SO 2 R 7 , optionally substituted (C 0 -C 4 alkylene)N(
  • each R 7 is independently selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 carbocyclyl, optionally substituted C 3-10 carbocyclylalkyl, optionally substituted C 2-6 heterocyclyl, optionally substituted C 2-10
  • heterocyclylalkyl or two R 11 groups together with the nitrogen to which they are attached join to form an optionally substituted C 2-6 N-heterocyclyl;
  • each R 8 is independently selected from H or optionally substituted C 1-6 alkyl
  • R 2 is absent or present, and when present is -H, -OH, optionally substituted C 1-6 alkyl, or halogen;
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • substituted or unsubstituted heteromonocycle substituted or unsubstituted heterobicycle, substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • U is C, CR 2 , or N;
  • is absent or present, and when present is a bond
  • each R 1 is independently halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, C 2-6 heterocyclyl, C 3-10 heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , (C 0 -C 4 alkylene)-CN, (C 0 -C 4 alkylene)-OR 7 , (C 0 -C 4 alkylene)-N(R 7 ) 2 , (C 0 -C 4 alkylene)N(R 8 )-COR 7 , (C 0 -C 4 alkylene)-SO 2 N(R 7 ) 2 , (C 0 -C 4 alkylene)-SO 2 R 7 , (C 0 -C 4 alkylene)N(R 8 )-SO 2 N(R 7 ) 2 , or (C 0 -C 4 alkylene)N(R 8 )-SO 2 R 7 ;
  • each R 7 is independently selected from H, C 1-6 alkyl, C 3-6 carbocyclyl, C 3-10
  • each R 8 is independently selected from H or C 1-6 alkyl
  • R 2 is absent or present, and when present is -H, -OH, C 1-6 alkyl, or halogen;
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • substituted or unsubstituted heteromonocycle substituted or unsubstituted heterobicycle, substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • Some embodiments provided herein describe a compound, or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, having the structure of Formula (I) wherein:
  • U is C, CR 2 , or N;
  • is absent or present, and when present is a bond
  • each R 1 is independently halogen, haloalkyl, or alkyl
  • R 2 is absent or present, and when present is -H, -OH, or halogen;
  • A is a bond
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • substituted or unsubstituted heteromonocycle substituted or unsubstituted heterobicycle, substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • U is CR 2 or N
  • is absent
  • each R 1 is independently Br, Cl, F, C 1-6 fluoroalkyl, or C 1-6 alkyl;
  • R 2 is absent or present, and when present is -H, -OH, Br, Cl, or F;
  • A is a bond
  • p 1, 2, or 3.
  • Some embodiments provided herein describe a compound, or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, for use in treating a metabolic disease or disorder, having the structure of Formula (II):
  • is absent or present, and when present is a bond
  • each R 1 is independently halogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-CN, optionally substituted (C 0 -C 4 alkylene)-OR 7 , optionally substituted (C 0 -C 4 alkylene)-N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 8 )-COR 7 , optionally substituted (C 0 -C 4 alkylene)- SO 2 N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-SO 2 R 7 , optionally substituted (C 0 - C 4 alkylene)N(R 8 )-SO 2 N(R 7 ) 2 , or optionally substituted (C 0
  • each R 7 is independently selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, or optionally substituted heteroarylalkyl; or two R 11 groups together with the nitrogen to which they are attached join to form an optionally substituted N-heterocyclyl;
  • each R 8 is independently selected from H or optionally substituted alkyl;
  • R 2 is absent or present, and when present is -H, -OH, optionally substituted alkyl, or halogen;
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • heterobicycle substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • Some embodiments provided herein describe a compound, or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, having the structure of Formula (II) wherein:
  • each R 1 is independently halogen, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 2-6 heterocyclyl, optionally substituted C 3-10 heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-CN, optionally substituted (C 0 -C 4 alkylene)-OR 7 , optionally substituted (C 0 -C 4 alkylene)- N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 8 )-COR 7 , optionally substituted (C 0 -C 4 alkylene)-SO 2 N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-SO 2 R 7 , optionally substituted (C 0 -C 4 alkylene)N(
  • each R 7 is independently selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 carbocyclyl, optionally substituted C 3-10 carbocyclylalkyl, optionally substituted C 2-6 heterocyclyl, optionally substituted C 2-10
  • heterocyclylalkyl or two R 11 groups together with the nitrogen to which they are attached join to form an optionally substituted C 2-6 N-heterocyclyl;
  • each R 8 is independently selected from H or optionally substituted C 1-6 alkyl
  • R 2 is absent or present, and when present is -H, -OH, optionally substituted C 1-6 alkyl, or halogen;
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • substituted or unsubstituted heteromonocycle substituted or unsubstituted heterobicycle, substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • is absent or present, and when present is a bond
  • each R 1 is independently halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, C 2-6 heterocyclyl, C 3-10 heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , (C 0 -C 4 alkylene)-CN, (C 0 -C 4 alkylene)-OR 7 , (C 0 -C 4 alkylene)-N(R 7 ) 2 , (C 0 -C 4 alkylene)N(R 8 )-COR 7 , (C 0 -C 4 alkylene)-SO 2 N(R 7 ) 2 , (C 0 -C 4 alkylene)-SO 2 R 7 , (C 0 -C 4 alkylene)N(R 8 )-SO 2 N(R 7 ) 2 , or (C 0 -C 4 alkylene)N(R 8 )-SO 2 R 7 ;
  • each R 7 is independently selected from H, C 1-6 alkyl, C 3-6 carbocyclyl, C 3-10
  • each R 8 is independently selected from H or C 1-6 alkyl
  • R 2 is absent or present, and when present is -H, -OH, C 1-6 alkyl, or halogen;
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • substituted or unsubstituted heteromonocycle substituted or unsubstituted heterobicycle, substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • Some embodiments provided herein describe a compound, or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, having the structure of Formula (II) wherein:
  • is absent or present, and when present is a bond
  • each R 1 is independently halogen, haloalkyl, or alkyl
  • R 2 is absent or present, and when present is -H, -OH, or halogen;
  • A is a bond
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • substituted or unsubstituted heteromonocycle substituted or unsubstituted heterobicycle, substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • is absent
  • each R 1 is independently Br, Cl, F, C 1-6 fluoroalkyl, or C 1-6 alkyl;
  • R 2 is -H, -OH, Br, Cl, or F;
  • A is a bond
  • p 1, 2, or 3.
  • is present and a bond, and R 2 is absent. In other embodiments, ⁇ is absent, and R 2 is present.
  • Some embodiments provided herein describe a compound, or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, for use in treating a metabolic disease or disorder, having the structure of Formula (III):
  • each R 1 is independently halogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-CN, optionally substituted (C 0 -C 4 alkylene)-OR 7 , optionally substituted (C 0 -C 4 alkylene)-N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 8 )-COR 7 , optionally substituted (C 0 -C 4 alkylene)- SO 2 N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-SO 2 R 7 , optionally substituted (C 0 - C 4 alkylene)N(R 8 )-SO 2 N(R 7 ) 2 , or optionally substituted (C 0
  • each R 7 is independently selected from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, or optionally substituted heteroarylalkyl; or two R 11 groups together with the nitrogen to which they are attached join to form an optionally substituted N-heterocyclyl;
  • each R 8 is independently selected from H or optionally substituted alkyl
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • heterobicycle substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • Some embodiments provided herein describe a compound, or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, having the structure of Formula (III) wherein:
  • each R 1 is independently halogen, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 2-6 heterocyclyl, optionally substituted C 3-10 heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-CN, optionally substituted (C 0 -C 4 alkylene)-OR 7 , optionally substituted (C 0 -C 4 alkylene)- N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 8 )-COR 7 , optionally substituted (C 0 -C 4 alkylene)-SO 2 N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-SO 2 R 7 , optionally substituted (C 0 -C 4 alkylene)N(R 8 )-SO 2 N(R 7 ) 2
  • each R 7 is independently selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 carbocyclyl, optionally substituted C 3-10 carbocyclylalkyl, optionally substituted C 2-6 heterocyclyl, optionally substituted C 2-10
  • heterocyclylalkyl or two R 11 groups together with the nitrogen to which they are attached join to form an optionally substituted C 2-6 N-heterocyclyl;
  • each R 8 is independently selected from H or optionally substituted C 1-6 alkyl
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • substituted or unsubstituted heteromonocycle substituted or unsubstituted heterobicycle, substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • each R 1 is independently halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, C 2-6 heterocyclyl, C 3-10 heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , (C 0 -C 4 alkylene)-CN, (C 0 -C 4 alkylene)-OR 7 , (C 0 -C 4 alkylene)-N(R 7 ) 2 , (C 0 -C 4 alkylene)N(R 8 )-COR 7 , (C 0 -C 4 alkylene)-SO 2 N(R 7 ) 2 , (C 0 -C 4 alkylene)-SO 2 R 7 , (C 0 -C 4 alkylene)N(R 8 )-SO 2 N(R 7 ) 2 , or (C 0 -C 4 alkylene)N(R 8 )-SO 2 R 7 ;
  • each R 7 is independently selected from H, C 1-6 alkyl, C 3-6 carbocyclyl, C 3-10
  • each R 8 is independently selected from H or C 1-6 alkyl
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • each R 1 is independently halogen, haloalkyl, or alkyl
  • A is a bond
  • B is substituted or unsubstituted monocycle, substituted or unsubstituted bicycle,
  • substituted or unsubstituted heteromonocycle substituted or unsubstituted heterobicycle, substituted or unsubstituted benzyl, -CO 2 H or -(C 1 -C 4 alkyl)CO 2 H; and p is 0, 1, 2, 3, 4, or 5.
  • each R 1 is independently Br, Cl, F, C 1-6 fluoroalkyl, or C 1-6 alkyl;
  • A is a bond
  • p 1, 2, or 3.
  • each R 1 is independently Br, Cl, F, C 1-6 fluoroalkyl, or C 1-6 alkyl;
  • p 1, 2, or 3.
  • each R 1 is independently halogen, haloalkyl, or alkyl
  • A is a bond
  • ⁇ , ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ are each independently absent or present, and when present each is a bond;
  • Z 1 is S, O, or N
  • Z 2 is S, O, N or NR 3 ,
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • X is C
  • R 4 is H, halogen, C 1 -C 10 alkyl, C 1 -C 10 cycloalkyl, -O(C 1 -C 10 alkyl), -C(O)OH, - C(O)O(C 1 -C 10 alkyl), -C(O)NH 2 , -C(O)NH(C 1 -C 4 alkyl), -C(O)N(C 1 -C 4 alkyl) 2 , -NHC(O)NH(C 1 -C 10 alkyl), -NHC(O)N(C 1 -C 4 alkyl) 2 , -SO 2 NH(C 1 -C 10 alkyl), -SO 2 N(C 1 -C l0 alkyl) 2 , -CN, or -CF 3 ;
  • R 5 is H or C 1 -C 10 alkyl
  • R 6 is H, C 1 -C 10 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 10 alkylene)CF 3 , -(C 1 - C 10
  • p 0, 1, 2, 3, 4, or 5.
  • each R 1 is independently halogen, haloalkyl, or alkyl
  • A is a bond
  • n 0;
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • Y 1 and Y 3 are each CH 2 or C(CH 3 ) 2 ;
  • Y 2 is O, SO 2 , or NR 6 ;
  • R 6 is H, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 4 alkylene)CF 3 , -(C 1 -C 4
  • alkylene)OCH 3 -(C 1 -C 4 alkylene)-halogen, -SO 2 (C 1 -C 4 alkyl), -SO 2 (C 1 -C 4 alkylene)CF 3 , -SO 2 (C 1 -C 4 alkylene)OCH 3 , -SO 2 (C 1 -C 4 alkylene)-halogen, - C(O)(C 1 -C 4 alkyl), -C(O)(C 1 -C 4 alkylene)CF 3 , -C(O)(C 1 -C 4 alkylene)OCH 3 , - C(O)(C 1 -C 4 alkylene)-halogen, -C(O)NH(C 1 -C 4 alkyl), -C(O)N(C 1 -C 4 alkyl) 2 , -(C 1 -C 4 alkylene)C(O)OH, -C(O)NH 2 , or oxetan
  • p 0, 1, 2, 3, 4, or 5.
  • is present, Z 1 is O or S, Z 2 is N, X is C, ⁇ is present, and ⁇ and ⁇ are absent.
  • is absent, then Z 1 is N, Z 2 is NR 3 , X is C, ⁇ and ⁇ are present, and ⁇ is absent.
  • is absent, then Z 1 is N, Z 2 is O or S, X is C, ⁇ and ⁇ are present, and ⁇ is absent.
  • each R 1 is independently halogen, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 cycloalkyl, optionally substituted C 2-6 heterocyclyl, optionally substituted C 3-10 heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-CN, optionally substituted (C 0 -C 4 alkylene)-OR 7 , optionally substituted (C 0 -C 4 alkylene)-N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 8 )-COR 7 , optionally substituted (C 0 -C 4 alkylene)- SO 2 N(R 7 ) 2 , optionally substituted (C 0 -C 4 alkylene)-SO 2 R 7 , optionally substituted (C 0 -C 4 alkylene)-SO 2 R 7 , optionally substituted (C 0
  • each R 1 is independently halogen, C 1-6 alkyl, C 1-6 haloalkyl, C 3-6 cycloalkyl, C 2-6 heterocyclyl, C 3-10 heterocyclalkyl, -COR 7 , -CON(R 7 ) 2 , (C 0 -C 4 alkylene)-CN, (C 0 -C 4 alkylene)-OR 7 , (C 0 -C 4 alkylene)-N(R 7 ) 2 , (C 0 -C 4 alkylene)N(R 8 )-COR 7 , (C 0 -C 4 alkylene)-SO 2 N(R 7 ) 2 , (C 0 -C 4 alkylene)-SO 2 R 7 , (C 0 -
  • each R 1 is independently halogen, C 1-6 alkyl, C 1-6 haloalkyl, -COR 7 , -CON(R 7 ) 2 , (C 0 - C 4 alkylene)-CN, (C 0 -C 4 alkylene)-OR 7 , or (C 0 -C 4 alkylene)-N(R 7 ) 2 .
  • each R 1 is independently (C 0 -C 4 alkylene)N(R 8 )-COR 7 , (C 0 -C 4 alkylene)-SO 2 N(R 7 ) 2 , (C 0 -C 4 alkylene)- SO 2 R 7 , (C 0 -C 4 alkylene)N(R 8 )-SO 2 N(R 7 ) 2 , or (C 0 -C 4 alkylene)N(R 8 )-SO 2 R 7 .
  • each R 1 is independently halogen, C 1-6 alkyl, C 1-6 haloalkyl, -COR 7 , -CON(R 7 ) 2 , -CN, (C 0 -C 4 alkylene)-OR 7 , or (C 0 -C 4 alkylene)-N(R 7 ) 2 .
  • each R 1 is independently halogen, C 1-6 alkyl, C 1-6 haloalkyl, or -CN.
  • each R 1 is independently F, Br, Cl, C 1-6 haloalkyl, or C 1-6 alkyl.
  • each R 1 is independently F or CF 3 .
  • each R 1 is -CF 3 .
  • each R 1 is independently selected from t-Bu, Cl, F, or–CF 3 .
  • each R 7 is independently selected from H, optionally substituted C 1-6 alkyl, optionally substituted C 3-6 carbocyclyl, optionally substituted C 3-10
  • carbocyclylalkyl optionally substituted C 2-6 heterocyclyl, optionally substituted C 2-10
  • each R 7 is independently selected from H, C 1-6 alkyl, C 3-6 carbocyclyl, C 3-10 carbocyclylalkyl, C 2-6 heterocyclyl, C 2-10 heterocyclylalkyl; or two R 11 groups together with the nitrogen to which they are attached join to form a C 2-6 N-heterocyclyl.
  • each R 7 is independently selected from H, C 1-6 alkyl, or C 3-6 carbocyclyl.
  • each R 7 is independently selected from H or C 1-6 alkyl. In some embodiments, each R 7 is H or Me.
  • each R 8 is independently selected from H, C 1-6 alkyl, or C 1-6 haloalkyl. In some embodiments, each R 8 is independently selected from H or C 1-6 alkyl. In some embodiments, each R 8 is independently selected from H or Me. In some embodiments, each R 8 is H.
  • p is 0, 1, 2, 3, or 4. In some embodiments, p is 0, 1, 2, or 3. In some embodiments, p is 0, 1 or 2. In some embodiments, p is 0 or 1. In some embodiments, p is 1, 2, or 3. In some embodiments, p is 1 or 2. In some embodiments, p is 1, 2, 3, or 4. In some embodiments, p is 2, 3, or 4. In some embodiments, p is 2 or 3. In some embodiments, p is 0. In some embodiments, p is 1. In some embodiments, p is 2. In some embodiments, p is 3. In some embodiments, p is 4. In some embodiments, p is 5. In some embodiments, p is 1.
  • R 2 is -H, -OH, optionally substituted alkyl, or halogen. In some embodiments, R 2 is -H, -OH, alkyl, haloalkyl, or halogen. In some embodiments, R 2 is -H, - OH, C 1-6 alkyl, C 1-6 haloalkyl, or halogen. In some embodiments, R 2 is -H, -OH, Me, CF 3 , or halogen. In some embodiments, R 2 is -H, -OH, Me, CF 3 , Cl, or F. In some embodiments, R 2 is -H, - OH, Me, CF 3 , or F.
  • R 2 is -H, -OH, or halogen. In some embodiments, R 2 is - H, -OH, or F. In some embodiments, R 2 is–H. In some embodiments, R 2 is–OH. In some embodiments, R 2 is F. In some embodiments, R 2 is Cl.
  • is absent or is present; each R 1 is independently selected from t-Bu, Cl, F, or–CF 3 ; and R 2 is absent or present, and when present is H, -OH, or F. In certain embodiments, ⁇ is absent; R 1 is–CF 3 ; and R 2 is H. In some embodiments, ⁇ is present; R 1 is–CF 3 ; and R 2 is absent.
  • B is a substituted or unsubstituted carbomonocycle. In some embodiments, B is substituted or unsubstituted 5 -, 6-, or 7-membered ring heteromonocycle containing one or more heteroatoms selected from N, O, or S.
  • B is a substituted or unsubstituted imidazole, substituted or unsubstituted pyridazine, substituted or unsubstituted triazine, substituted or unsubstituted pyrazole, substituted or unsubstituted pyridine, substituted or unsubstituted pyrimidine, substituted or unsubstituted pyrazine, substituted or unsubstituted thiadiazole, substituted or unsubstituted thiazole, substituted or unsubstituted pyrrole, substituted or unsubstituted oxazole, or substituted or unsubstituted triazole.
  • B is a substituted or unsubstituted imidazole, substituted or unsubstituted pyrazole, substituted or unsubstituted thiadiazole, substituted or unsubstituted thiazole, substituted or unsubstituted pyrrole, substituted or unsubstituted oxazole, or substituted or unsubstituted triazole.
  • B is a substituted or unsubstituted pyridazine, substituted or unsubstituted triazine, substituted or unsubstituted pyridine, substituted or unsubstituted pyrimidine, or substituted or unsubstituted pyrazine.
  • B is a substituted or unsubstituted pyridine, substituted or unsubstituted pyrimidine, or substituted or unsubstituted pyrazine. In certain embodiments, B is a substituted or unsubstituted pyridine. In some embodiments, B is a substituted or unsubstituted pyrimidine. In some embodiments, B is a substituted or unsubstituted pyrazine.
  • B has the structure:
  • each R 3 is independently optionally substituted alkyl, optionally substituted aryl, optionally substituted aralkyl, optionally substituted heteroaryl, optionally substituted heteroaralkyl, optionally substituted heterocyclyl, optionally substituted heterocyclalkyl, -COR 13 , -CON(R 13 ) 2 , optionally substituted (C 0 -C 4 alkylene)-CN, optionally substituted (C 0 -C 4 alkylene)-CO(O)H, optionally substituted (C 0 -C 4 alkylene)-OR 13 , optionally substituted (C 0 -C 4 alkylene)-N(R 13 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 14 )-COR 13 , optionally substituted (C 0 -C 4 alkylene)N(R 14 )-CO 2 R 13 , optionally substituted (C 0 -C 4 alkylene)N(R 14 )
  • each R 14 is independently selected from H or optionally substituted alkyl; and m is 0, 1, 2, or 3.
  • each R 3 is independently optionally substituted alkyl, -COR 13 , -CON(R 13 ) 2 , optionally substituted (C 0 -C 4 alkylene)-CN, optionally substituted (C 0 -C 4 alkylene)- CO(O)H, optionally substituted (C 0 -C 4 alkylene)-OR 13 , optionally substituted (C 0 -C 4 alkylene)- N(R 13 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 14 )-COR 13 , optionally substituted (C 0 -C 4 alkylene)N(R 14 )-CO 2 R 13 , optionally substituted (C 0 -C 4 alkylene)N(R 14 )-CON(R 13 ) 2 , optionally substituted (C 0 -C 4 alkylene)N(R 14 )-SO 2 R 13 , optionally substituted (C 0 -C 4 alkylene)
  • each R 3 is independently alkyl, -COR 13 , -CON(R 13 ) 2 , (C 0 -C 4 alkylene)-CN, (C 0 -C 4 alkylene)-CO(O)H, (C 0 -C 4 alkylene)-OR 13 , (C 0 -C 4 alkylene)-N(R 13 ) 2 , (C 0 -C 4 alkylene)N(R 14 )-COR 13 , (C 0 -C 4 alkylene)N(R 14 )-CO 2 R 13 , (C 0 -C 4 alkylene)N(R 14 )-CON(R 13 ) 2 , (C 0 -C 4 alkylene)N(R 14 )-SO 2 R 13 , (C 0 -C 4 alkylene)-SO 2 N(R 13 ) 2 , or (C 0 - C 4 alkylene)-SO 2 R 13 .
  • each R 3 is independently alkyl, -COR 13 , -CON(R 13 ) 2 , (C 0 -C 4 alkylene)-CN, (C 0 -C 4 alkylene)-CO(O)H, (C 0 -C 4 alkylene)-OR 13 , (C 0 -C 4 alkylene)-N(R 13 ) 2 , (C 0 -C 4 alkylene)N(R 14 )-COR 13 , (C 0 -C 4 alkylene)N(R 14 )-CO 2 R 13 , (C 0 -C 4 alkylene)N(R 14 )-CON(R 13 ) 2 , (C 0 -C 4 alkylene)N(R 14 )-SO 2 R 13 , (C 0 -C 4 alkylene)-SO 2 N(R 13 ) 2 , or (C 0 -C 4 alkylene)-SO 2 R 13 .
  • each R 13 is independently selected from H, alkyl, haloalkyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl; or two R 13 groups together with the nitrogen to which they are attached join to form an optionally substituted N-heterocyclyl.
  • each R 13 is independently selected from H, alkyl, or haloalkyl. In some embodiments, each R 13 is independently selected from H or alkyl. In further or additional embodiments, each R 14 is independently selected from H or alkyl.
  • each R 3 is independently halogen, -CF 3 , -OCH 3 , SO 2 CH 3 , - OH, NH 2 , C 1 -C 4 alkyl , C 3 -C 6 cycloalkyl, -O(C 1 -C 4 alkyl), -C(O)NH 2 , -C(O)NH(C 1 -C 10 alkyl), - C(O)N(C 1 -C 10 alkyl) 2 , -C(O)OH, -C(O)O(C 1 -C 4 alkyl), -C(O)(C 1 -C 4 alkyl), -C(O)NHSO 2 (C 1 -C 10 alkyl), -C(O)NH(C 3 -C 6 cycloalkyl), -C(O)NH(aryl), -OSO 2 NH 2 , -NHC(O)NH(C 1 -C 10 alkyl, C 3 -C
  • B has the structure: , B has the
  • B has the structure:
  • ⁇ , ⁇ , ⁇ , and ⁇ are each independently absent or present, and when present each is a bond;
  • X is C
  • Z 1 is S, O, or N
  • Z 2 is S, O, N, or NR 3 ;
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • C is a substituted or unsubstituted fused 5-, 6-, or 7- membered ring structure.
  • is present, Z 1 is O or S, Z 2 is N, X is C, ⁇ is present, and ⁇ and ⁇ are absent.
  • is absent, Z 1 is N, Z 2 is NR 3 , X is C, ⁇ and ⁇ are present, and ⁇ is absent.
  • is absent, Z 1 is N, Z 2 is O or S, X is C, ⁇ and ⁇ are present, and ⁇ is absent.
  • B has the structure:
  • n 0, 1, or 2;
  • ⁇ , ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ are each independently absent or present, and when present each is a bond;
  • Z 1 is S, O, or N
  • Z 2 is S, O, N or NR 3 ,
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • X is C
  • R 4 is H, halogen, C 1 -C 10 alkyl, C 1 -C 10 cycloalkyl, -O(C 1 -C 10 alkyl), -C(O)OH, - C(O)O(C 1 -C 10 alkyl), -C(O)NH 2 , -C(O)NH(C 1 -C 4 alkyl), -C(O)N(C 1 -C 4 alkyl) 2 , - NHC(O)NH(C 1 -C 10 alkyl), -NHC(O)N(C 1 -C 4 alkyl) 2 , -SO 2 NH(C 1 -C 10 alkyl), - SO 2 N(C 1 -C l0 alkyl) 2 , -CN, or -CF 3 ;
  • R 5 is H or C 1 -C 10 alkyl
  • R 6 is H, C 1 -C 10 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 10 alkylene)CF 3 , -(C 1 - C 10 alkylene)OCH 3 , -(C 1 -C 10 alkylene)-halogen, -SO 2 (C 1 -C 10 alkyl), -SO 2 (C 1 -C 10 alkylene) -CF 3 , -SO 2 (C 1 -C 10 alkylene)OCH 3 , -SO 2 (C 1 -C 10 alkylene)-halogen, - C(O)(C 1 -C 10 alkyl), -C(O)(C 1 -C 10 alkylene)CF 3 , -C(O)(C 1 -C 10 alkylene)OCH 3 , - C(O)(C 1 -C 10 alkylene)-halogen, -C(O)NH(C 1 -C 10 alkyl),
  • is present, Z 1 is O or S, Z 2 is N, X is C, ⁇ is present, and ⁇ and ⁇ are absent.
  • is absent, Z 1 is N, Z 2 is N, X is C, ⁇ and ⁇ are present, and ⁇ is absent.
  • is absent, Z 1 is N, Z 2 is O or S, X is C, ⁇ and ⁇ are present, and ⁇ is absent.
  • ⁇ and ⁇ are present. In some embodiments, ⁇ , ⁇ , ⁇ , and ⁇ are absent. In other embodiments, ⁇ , ⁇ , ⁇ , and ⁇ are present. In certain embodiments, ⁇ , and ⁇ are absent.
  • Z 1 is N.
  • Z 2 is O, S, or NR 3 ; and R 3 is H, C 1 -C 4 alkyl, or oxetane. In other embodiments, Z 2 is O or NR 3 ; R 3 is H, C 1 -C 4 alkyl, or oxetane.
  • X is C.
  • ⁇ and ⁇ are present; ⁇ , ⁇ , ⁇ , and ⁇ are absent; Z 1 is N; Z 2 is O, S, or NR 3 ; R 3 is H, C 1 -C 4 alkyl, or oxetane; and X is C.
  • ⁇ , ⁇ , ⁇ , and ⁇ are present; ⁇ , and ⁇ are absent; Z 1 is N; Z 2 is O or NR 3 ; R 3 is H, C 1 -C 4 alkyl, or oxetane; and X is C.
  • B has the structure:
  • n 0;
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • Y 1 and Y 3 are each CH 2 or C(CH 3 ) 2 ;
  • Y 2 is O, SO 2 , or NR 6 ;
  • R 6 is H, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 4 alkylene)CF 3 , -(C 1 -C 4 alkylene)OCH 3 , -(C 1 -C 4 alkylene)-halogen, -SO 2 (C 1 -C 4 alkyl), -SO 2 (C 1 -C 4 alkylene)CF 3 , -SO 2 (C 1 - C 4 alkylene)OCH 3 , -SO 2 (C 1 -C 4 alkylene)-halogen, -C(O)(C 1 -C 4 alkyl), -C(O)(C 1 - C 4 alkylene)CF 3 , -C(O)(C 1 -C 4 alkylene)OCH 3 , -C(O)(C 1 -C 4 alkylene)-halogen, - C(O)NH(C 1 -C 4 alkyl), -
  • n 1;
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • Y 1 and Y 4 are CH 2 or C(CH 3 ) 2 ;
  • Y 2 and Y 3 are each CH 2 or C(CH 3 ) 2 , O, SO 2 , or NR 6 ;
  • R 6 is H, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 4 alkylene)CF 3 , -(C 1 -C 4 alkylene)OCH 3 , -(C 1 -C 4 alkylene)-halogen, -SO 2 (C 1 -C 4 alkyl), -SO 2 (C 1 -C 4 alkylene)CF 3 , -SO 2 (C 1 -C 4 alkylene)OCH 3 , -SO 2 (C 1 -C 4 alkylene)-halogen, -C(O)(C 1 -C 4 alkyl), -C(O)(C 1 -C 4 alkylene)CF 3 , -C(O)(C 1 -C 4 alkylene)OCH 3 , -C(O)(C 1 -C 4 alkylene)-halogen, - C(O)NH(C 1 -C 4 alkyl), -
  • n 2;
  • R 3 is H, C 1 -C 4 alkyl, or oxetane
  • Y 1 and Y 4 are CH 2 or C(CH 3 ) 2 ;
  • Y 2 and Y 3 are each CH 2 or C(CH 3 ) 2 , O, SO 2 , or NR 6 ;
  • R 6 is H, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 4 alkylene)CF 3 , -(C 1 -C 4 alkylene)OCH 3 , -(C 1 -C 4 alkylene)-halogen, -SO 2 (C 1 -C 4 alkyl), -SO 2 (C 1 -C 4 alkylene)CF 3 , -SO 2 (C 1 -C 4 alkylene)OCH 3 , -SO 2 (C 1 -C 4 alkylene)-halogen, -C(O)(C 1 -C 4 alkyl), -C(O)(C 1 -C 4 alkylene)CF 3 , -C(O)(C 1 -C 4 alkylene)OCH 3 , -C(O)(C 1 -C 4 alkylene)-halogen, - C(O)NH(C 1 -C 4 alkyl), -
  • n is 0 or 1. In some embodiments, n is 1 or 2. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2.
  • B has the structure:
  • B has the structure In other embodiments, B has the structure
  • B has the structure
  • B has the structure . In some embodiments, B has the structure
  • B has the structure:
  • B has the structure .
  • B has the structure . In some embodiments, B has the structure
  • B has the structure . In some embodiments,
  • B has the structure In some embodiments, B has the structure
  • B has the structure In some embodiments, B has the structure
  • B has the structure
  • B has the structure:
  • B has the structure . In some embodiments, B has the structure
  • B has the structure . In some embodiments, B
  • B has the structure .
  • B has the structure
  • R 6 is H, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 6 alkylene)CF 3 , - (C 1 - C 6 alkylene)OCH 3 , -(C 1 -C 6 alkylene)-halogen, -SO 2 -C 1 -C 6 alkyl, -SO 2 (C 1 -C 6 alkylene)-CF 3 , - SO 2 (C 1 -C 6 alkylene)OCH 3 , -SO 2 (C 1 -C 6 alkylene)-halogen, -C(O)(C 1 -C 6 alkyl), -C(O)(C 1 -C 6 alkylene)CF 3 , -C(O)(C 1 -C 6 alkylene)OCH 3 , -C(O)(C 1 -C 6 alkylene)-halogen, -C(O)NH(C 1
  • R 6 is C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, -(C 1 -C 6 alkylene)CF 3 , -(C 1 - C 6 alkylene)OCH 3 , -(C 1 -C 6 alkylene)- halogen, -SO 2 -C 1 -C 6 alkyl, -SO 2 (C 1 -C 6 alkylene)-CF 3 , -SO 2 (C 1 -C 6 alkylene)OCH 3 , -SO 2 (C 1 -C 6 alkylene)-halogen, -C(O)(C 1 -C 6 alkyl), -C(O)(C 1 -C 6 alkylene)CF 3 , -C(O)(C 1 -C 6 alkylene)OCH 3 , - C(O)(C 1 -C 6 alkylene)-halogen, -C(O)NH(C 1 -C 6 alkyl,
  • R 6 is -C(O)(C 1 -C 6 alkyl).
  • R 6 is H, C 1 -C 4 alkyl, -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 CH(CH 3 ) 2 , t-Bu, -CH 2 OCH 3 , - CH 2 CF 3 , -CH 2 Cl, -CH 2 F, -CH 2 CH 2 OCH 3 , -CH 2 CH 2 CF 3 , -CH 2 CH 2 Cl, -CH 2 CH 2 F, , - SO 2 CH 3 , -SO 2 CH 2 CH 3 , -SO 2 CH 2 CH 3 , -SO 2 CH(CH 3 ) 2 , -SO 2 CH 2 CH(CH 3 ) 2 , -SO 2 (t-Bu), - SO 2 CH 3 , -SO 2 CH 2 CH 3 , -SO 2 CH(CH 3 ) 2 , -SO 2 CH 2 CH(CH 3 )
  • R 6 is -C(O)(C 1 -C 6 alkyl). In some embodiments, R 6 is H, C 1 -C 4 alkyl, -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -CH 2 CH(CH 3 ) 2 , t-Bu, -CH 2 OCH 3 , - CH 2 CF 3 , -CH 2 Cl, -CH 2 F, -CH 2 CH 2 OCH 3 , -CH 2 CH 2 CF 3 , -CH 2 CH 2 Cl, -CH 2 CH 2 F, or In other embodiments, R 6 is -SO 2 CH 3 , -SO 2 CH 2 CH 3 , -SO 2 CH 2 CH 2 CH 3 , -SO 2 CH(CH 3 ) 2 , - SO 2 CH 2 CH(CH 3 ) 2 , -SO 2 (t-Bu), -SO 2 CH 2 OCH 3 , -SO 2 CH 2 CF 3 , -SO 2 CH
  • R 6 is C(O)CH 3 , C(O)CH 2 CH 3 , -C(O)CH 2 CH 2 CH 3 , -C(O)CH(CH 3 ) 2 , - C(O)CH 2 CH(CH 3 ) 2 , -C(O)t-Bu, -C(O)CH 2 OCH 3 , -C(O)CH 2 CF 3 , -C(O)CH 2 Cl, -C(O)CH 2 F, -
  • B has the structure:
  • B has the structure
  • B has the structure . In some embodiments, B has the structure . In some embodiments, B has the structure . In some embodiments, B has the structure In
  • B has the structure In some embodiments, B has the structure
  • B has the structure . In some embodiments,
  • B has the structure In some embodiments, B has the structure
  • B has the structure:
  • Y 1 , Y 2 , Y 3 and each occurrence of Y 4 are each independently CR 4 , or N; wherein:
  • R 3 is H, halogen, C 1 -C 10 alkyl, C 1 -C 10 cycloalkyl , -O(C 1 -C 10 alkyl), -C(O)OH, - C(O)O(C 1 -C 10 alkyl), -C(O)NH 2 , -C(O)NH(C 1 -C 4 alkyl), -C(O)N(C 1 -C 4 alkyl) 2 , - NHC(O)NH(C 1 -C 10 alkyl), -NHC(O)N(C 1 -C 4 alkyl) 2 , -SO 2 NH(C 1 -C 10 alkyl), - SO 2 N(C 1 -C l0 alkyl) 2 , -CN, or -CF 3 .
  • Y 1 , Y 2 , Y 3 and Y 4 are CH. In other embodiments, Y 1 , Y 2 , Y 3 are CH and Y 4 is N. In some embodiments, Y 1 , Y 2 , Y 4 are CH and Y 3 is N. In some embodiments, Y 1 , Y 3 , Y 4 are CH and Y 2 is N. In certain embodiments, Y 2 , Y 3 , Y 4 are CH and Y 1 is N.
  • B has the structure:
  • R 3 is H, halogen, C 1 -C 10 alkyl, C 1 -C 10 cycloalkyl , -O(C 1 -C 10 alkyl), -C(O)OH, - C(O)O(C 1 -C 10 alkyl), -C(O)NH 2 , -C(O)NH(C 1 -C 4 alkyl), -C(O)N(C 1 -C 4 alkyl) 2 , - NHC(O)NH(C 1 -C 10 alkyl), -NHC(O)N(C 1 -C 4 alkyl) 2 , -SO 2 NH(C 1 -C 10 alkyl), - SO 2 N(C 1 -C l0 alkyl) 2 , -CN, or -CF 3 ; and
  • R 4 is H, halogen, C 1 -C 4 alkyl, C 3 -C 6 cycloalkyl, -O(C 1 -C 4 alkyl), -CN, -CF 3 , - C(O)OH, -C(O)NH 2 , -C(O)N(CH 3 ) 2 , -C(O)NHCH 3 , or -NHC(O)N(CH 3 ) 2 .
  • B has the structure . In certain embodiments,
  • B has the structure . In some embodiments, B has the structure
  • B has the structure .
  • each R 4 is independently H, halo, optionally substituted alkyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, or optionally substituted heterocyclylalkyl. In some embodiments, each R 4 is independently H, halo, optionally substituted alkyl, or optionally substituted carbocyclyl. In some embodiments, each R 4 is independently halogen, haloalkyl, or alkyl. In certain embodiments, each R 1 is independently F, Br, Cl, C 1-6 haloalkyl, or C 1-6 alkyl.
  • a compound of Formula (I) or (II) has the structure:
  • a compound of Formula (I) or (II) has the structure:
  • a compound of Formula (I) or (II) has the
  • heterocyclic compounds of Formula (I), (II), or (III) are provided in Table 1. TABLE 1
  • Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation include for example, “Synthetic Organic Chemistry”, John Wiley & Sons, Inc., New York; S. R. Sandler et al., “Organic Functional Group Preparations,” 2nd Ed., Academic Press, New York, 1983; H. O. House, “Modern Synthetic Reactions", 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L. Gilchrist, "Heterocyclic Chemistry", 2nd Ed., John Wiley & Sons, New York, 1992; J. March, “Advanced Organic Chemistry: Reactions, Mechanisms and Structure", 4th Ed.,
  • Retinol-binding protein 4 (RBP4), the sole retinol transporter in blood, is secreted from adipocytes and liver. Serum RBP4 levels correlate highly with insulin resistance, other metabolic syndrome factors, and cardiovascular disease. In some instances, elevated serum RBP4 causes insulin resistance and impaired glucose tolerance. In some instances, lowering of serum RBP4 improves insulin action and glucose tolerance. In some embodiments, the compounds described herein lower serum or plasma RBP4 and thus improve insulin action and glucose tolerance.
  • the serum or plasma levels of RBP4 are reduced by at least 50% from baseline.
  • 48 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 65% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 80% from baseline. In some embodiments, 48 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 85% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 65% from baseline.
  • 36 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 80% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 85% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 30% from baseline. In some embodiments, 24 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 40% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 65% from baseline.
  • 24 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 80% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 85% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 30% from baseline. In some embodiments, 12 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 40% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 65% from baseline.
  • 12 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 80% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 85% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 20% from baseline. In some embodiments, 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 25% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 30% from baseline. In some embodiments, 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 30% from baseline. In some embodiments, 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a
  • the serum or plasma levels of RBP4 are reduced by at least 40% from baseline.
  • 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 50% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 65% from baseline. In certain embodiments, 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 80% from baseline. In some embodiments, 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a
  • the serum or plasma levels of RBP4 are reduced by at least 85% from baseline.
  • the serum or plasma levels of RBP4 are reduced by at least 1 mg/dL. In other embodiments, 48 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 2 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 10 mg/dL.
  • 48 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 15 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 10 mg/dL.
  • 36 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 15 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 1 mg/dL. In other embodiments, 24 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 2 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 5 mg/dL. In certain embodiments, 24 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 5 mg/dL. In certain embodiments, 24 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 5 mg/dL. In certain embodiments, 24 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereo
  • the serum or plasma levels of RBP4 are reduced by at least 10 mg/dL.
  • 24 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 15 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 1 mg/dL. In other embodiments, 12 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 2 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 10 mg/dL.
  • 12 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 15 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 1 mg/dL. In other embodiments, 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 2 mg/dL.
  • the serum or plasma levels of RBP4 are reduced by at least 5 mg/dL. In certain embodiments, 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 5 mg/dL. In certain embodiments, 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof, the serum or plasma levels of RBP4 are reduced by at least 5 mg/dL. In certain embodiments, 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereo
  • the serum or plasma levels of RBP4 are reduced by at least 10 mg/dL.
  • 6 hours after administration of a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof the serum or plasma levels of RBP4 are reduced by at least 15 mg/dL.
  • a compound disclosed herein is used to treat or ameliorate a disease associated with altered RBP4 pathways when administered to a subject in need thereof.
  • a compound disclosed herein is used to treat or ameliorate the effects of a disease associated with altered RBP4 pathway when administered to a subject in need thereof.
  • Exemplary diseases associated with altered RBP4 include metabolic diseases.
  • a compound disclosed herein is used to treat or ameliorate a metabolic disease when administered to a subject in need thereof.
  • Exemplary metabolic diseases include NASH, NAFLD, type II diabetes, obesity, fibrosis, cirrhosis, or liver cancer.
  • the fatty liver disease is selected from the group consisting of non-alcoholic fatty acid liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), fatty liver disease resulting from hepatitis, fatty liver disease resulting from obesity, fatty liver disease resulting from diabetes, fatty liver disease resulting from insulin resistance, fatty liver disease resulting from hypertriglyceridemia, Abetalipoproteinemia, glycogen storage diseases, Weber- Christian disease, Wolmans disease, acute fatty liver of pregnancy, and lipodystrophy.
  • NAFLD non-alcoholic fatty acid liver disease
  • NASH non-alcoholic steatohepatitis
  • fatty liver disease resulting from obesity fatty liver disease resulting from diabetes
  • fatty liver disease resulting from insulin resistance fatty liver disease resulting from hypertriglyceridemia
  • Abetalipoproteinemia glycogen storage diseases
  • Weber- Christian disease Wolmans disease
  • Wolmans disease acute fatty liver of pregnancy, and lipodystrophy
  • NASH Non-Alcoholic Steatohepatitis
  • Non-alcoholic steatohepatitis or NASH is a common liver disease, which resembles alcoholic liver disease, but occurs in people who drink little or no alcohol.
  • the major feature in NASH is fat in the liver, along with inflammation and damage.
  • NASH progresses into advanced NASH, which is characterized, inter cilia, by hepatic fibrosis.
  • NASH progresses to cirrhosis, in which the liver is damaged, scarred, and is no longer able to work properly.
  • a person with cirrhosis experiences fluid retention, muscle wasting, bleeding from the intestines, and/or liver failure.
  • NASH is associated with an increased risk of cardiovascular mortality and type II diabetes mellitus. Cirrhosis due to NASH increases the risk of hepatocellular carcinoma.
  • NASH is usually first suspected in a person who is found to have elevations in liver tests that are included in routine blood test panels, such as alanine aminotransferase (ALT) or aspartate aminotransferase (AST). When further evaluation shows no apparent reason for liver disease (such as medications, viral hepatitis, or excessive use of alcohol) and when X-rays or imaging studies of the liver show fat, NASH is suspected.
  • NASH is diagnosed and separated from NAFLD by a liver biopsy. For a liver biopsy, a needle is inserted through the skin to remove a small piece of the liver. NASH is diagnosed when examination of the tissue with a microscope shows fat along with inflammation and damage to liver cells. A biopsy can provide information about scar tissue has development in the liver.
  • heterocyclic RBP4 inhibitory compounds described herein for treating NASH in a subject in need thereof.
  • the heterocyclic RBP4 inhibitory compounds inhibit NASH.
  • the heterocyclic RBP4 inhibitory compounds arrest the development of NASH or its clinical symptoms.
  • the heterocyclic RBP4 inhibitory compounds reduce the development of NASH or its clinical symptoms.
  • the heterocyclic RBP4 inhibitory compounds relieve the subject of NASH.
  • the heterocyclic RBP4 inhibitory compounds cause regression, reversal or amelioration of NASH.
  • the heterocyclic RBP4 inhibitory compounds reduce the number, frequency, duration, or severity of NASH clinical symptoms.
  • heterocyclic RBP4 inhibitory compounds are used
  • the heterocyclic RBP4 inhibitory compounds are used to prevent or reduce the risk of developing NASH.
  • the heterocyclic RBP4 inhibitory compounds cause the clinical symptoms of NASH not to develop in a subject who may be predisposed to NASH but who does not yet experience or display symptoms of NASH.
  • NAFLD Non-Alcoholic Fatty Liver Disease
  • Non-alcoholic fatty liver disease is a disorder affecting as many as 1 in 3-5 adults and 1 in 10 children in the United States, and refers to conditions where there is an accumulation of excess fat in the liver of people who drink little or no alcohol.
  • the most common form of NAFLD is a non-serious condition called hepatic steatosis (fatty liver), in which fat accumulates in the liver cells.
  • NAFLD most often presents itself in individuals with a constellation of risk factors called the metabolic syndrome, which is characterized by elevated fasting plasma glucose (FPG) with or without intolerance to post-prandial glucose, being overweight or obese, high blood lipids such as cholesterol and triglycerides (TGs) and low high-density lipoprotein cholesterol (HDL-C) levels, and high blood pressure; but not all patients have all the manifestations of the metabolic syndrome.
  • FPG fasting plasma glucose
  • TGs cholesterol and triglycerides
  • HDL-C low high-density lipoprotein cholesterol
  • NAFLD Newcastle disease virus
  • the diagnosis of NAFLD is usually first suspected in an overweight or obese person who is found to have mild elevations in their liver blood tests during routine testing, though NAFLD can be present with normal liver blood tests, or incidentally detected on imaging investigations such as abdominal ultrasound or CT scan. It is confirmed by imaging studies, most commonly a liver ultrasound or magnetic resonance imaging (MRI), and exclusion of other causes.
  • MRI magnetic resonance imaging
  • Some embodiments provided herein describe the use of the heterocyclic RBP4 inhibitory compounds described herein for treating NAFLD in a subject in need thereof.
  • the heterocyclic RBP4 inhibitory compounds inhibit NAFLD. In specific embodiments, the heterocyclic RBP4 inhibitory compounds arrest the development of NAFLD or its clinical symptoms. In other embodiments, the heterocyclic RBP4 inhibitory compounds reduce the development of NAFLD or its clinical symptoms. In certain embodiments, the heterocyclic RBP4 inhibitory compounds relieve the subject of NAFLD. In specific embodiments, the heterocyclic RBP4 inhibitory compounds cause regression, reversal or amelioration of NAFLD. In certain specific embodiments, the heterocyclic RBP4 inhibitory compounds reduce the number, frequency, duration, or severity of NAFLD clinical symptoms.
  • heterocyclic RBP4 inhibitory compounds are used
  • the heterocyclic RBP4 inhibitory compounds are used to prevent or reduce the risk of developing NAFLD.
  • the heterocyclic RBP4 inhibitory compounds cause the clinical symptoms of NAFLD not to develop in a subject who may be predisposed to NAFLD but who does not yet experience or display symptoms of NAFLD.
  • Type II diabetes is a metabolic disorder characterized by hyperglycemia and abnormities in the glucose-protein- and lipid-metabolism. Type II diabetes is caused by insulin resistance which is not adequately compensated due to an insufficient ⁇ -cell secretory capacity. In recent years, type II diabetes and its chronic complications have become a major threat to human health.
  • Type II diabetes results in the damage of many tissues and organs, which in turn leads to a variety of diabetic chronic complications, such as coronary heart disease, atherosclerosis, cerebrovascular disease and other diabetic macrovascular diseases, diabetic nephropathy, diabetic retinopathy and other diabetic microangiopathy, diabetic neuropathy, diabetic foot, diabetic maculopathy, diabetic cataract, diabetic glaucoma, refractive changes, iris and ciliary body diseases.
  • diabetic chronic complications such as coronary heart disease, atherosclerosis, cerebrovascular disease and other diabetic macrovascular diseases, diabetic nephropathy, diabetic retinopathy and other diabetic microangiopathy, diabetic neuropathy, diabetic foot, diabetic maculopathy, diabetic cataract, diabetic glaucoma, refractive changes, iris and ciliary body diseases.
  • heterocyclic RBP4 inhibitory compounds described herein for treating type II diabetes, including its chronic complications, in a subject in need thereof.
  • the heterocyclic RBP4 inhibitory compounds inhibit type II diabetes, including its chronic complications.
  • the heterocyclic RBP4 inhibitory compounds arrest the development of type II diabetes, its chronic complications, or its clinical symptoms.
  • the heterocyclic RBP4 inhibitory compounds reduce the development of type II diabetes, its chronic complications, or its clinical symptoms.
  • the heterocyclic RBP4 inhibitory compounds relieve the subject of type II diabetes, its chronic complications, or its clinical symptoms.
  • the heterocyclic RBP4 inhibitory compounds cause regression, reversal, or amelioration of type II diabetes, its chronic complications, or its clinical symptoms. In certain specific embodiments, the heterocyclic RBP4 inhibitory compounds reduce the number, frequency, duration, or severity of type II diabetes, its chronic complications, or its clinical symptoms.
  • heterocyclic RBP4 inhibitory compounds are used
  • the heterocyclic RBP4 inhibitory compounds are used to prevent or reduce the risk of type II diabetes, its chronic complications, or its clinical symptoms.
  • the heterocyclic RBP4 inhibitory compounds cause the clinical symptoms of type II diabetes not to develop in a subject who may be predisposed to type II diabetes but who does not yet experience or display symptoms of type II diabetes.
  • the subject is genetically, environmentally, dietarily or socially predisposed to type II diabetes.
  • Obesity and disorders associated with obesity such as diabetes are a major global health concern.
  • Obesity which is generally associated with an abnormal accumulation of fat cells, develops when energy intake exceeds energy expenditure.
  • Obesity is associated with an increased risk of diabetes. Most obese people are insulin resistant and have to adapt by increasing insulin secretion. In some instances, type II diabetes mellitus manifests in individuals who lose the ability to produce sufficient quantities of insulin to maintain normoglycemia in the face of insulin resistance.
  • Obesity is a state of chronic, low-grade inflammation, and macrophages are thought to play an important role in maintaining this state in adipose tissue. Many molecules secreted by adipose tissue promote adipose tissue inflammation. Emerging evidence suggests a possible role for proinflammatory pathways in RBP4-induced insulin resistance. RBP4 expression in adipose tissue and serum RBP4 levels strongly correlate with subclinical inflammation, including serum levels and adipose tissue expression of proinflammatory cytokines.
  • the heterocyclic RBP4 inhibitory compounds arrest the development of obesity or its clinical symptoms. In other embodiments, the heterocyclic RBP4 inhibitory compounds reduce the development of obesity or its clinical symptoms. In certain embodiments, the heterocyclic RBP4 inhibitory compounds relieve the subject of obesity. In specific embodiments, the heterocyclic RBP4 inhibitory compounds cause regression, reversal or amelioration of obesity. In certain specific embodiments, the heterocyclic RBP4 inhibitory compounds reduce the number, frequency, duration, or severity of obesity clinical symptoms.
  • heterocyclic RBP4 inhibitory compounds are used
  • the heterocyclic RBP4 inhibitory compounds are used to prevent or reduce the risk of developing obesity.
  • the heterocyclic RBP4 inhibitory compounds cause the clinical symptoms of obesity not to develop in a subject who may be predisposed to obesity but who does not yet experience or display symptoms of obesity.
  • the heterocyclic RBP4 inhibitory compound as described herein is administered as a pure chemical.
  • the heterocyclic RBP4 inhibitory compound described herein is combined with a pharmaceutically suitable or acceptable carrier (also referred to herein as a pharmaceutically suitable (or acceptable) excipient, physiologically suitable (or acceptable) excipient, or physiologically suitable (or acceptable) carrier) selected on the basis of a chosen route of administration and standard pharmaceutical practice as described, for example, in Remington: The Science and Practice of Pharmacy (Gennaro, 21 st Ed. Mack Pub. Co., Easton, PA (2005)).
  • composition comprising at least one heterocyclic RBP4 inhibitory compound, or a stereoisomer, pharmaceutically acceptable salt, or N-oxide thereof, together with one or more pharmaceutically acceptable carriers.
  • the carrier(s) or excipient(s) is acceptable or suitable if the carrier is compatible with the other ingredients of the composition and not deleterious to the recipient (i.e., the subject or patient) of the composition.
  • One embodiment provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • the pharmaceutical compositions are provided in a dosage form for oral administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers.
  • the heterocyclic RBP4 inhibitory compound as described by Formula (I), Formula (II), or Formula (III) is substantially pure, in that it contains less than about 5%, or less than about 1%, or less than about 0.1%, of other organic small molecules, such as unreacted intermediates or synthesis by-products that are created, for example, in one or more of the steps of a synthesis method.
  • Suitable oral dosage forms include, for example, tablets, pills, sachets, or capsules of hard or soft gelatin, methylcellulose or of another suitable material easily dissolved in the digestive tract.
  • suitable nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. (See, e.g., Remington: The Science and Practice of Pharmacy (Gennaro, 21 st Ed. Mack Pub. Co., Easton, PA (2005)).
  • the pharmaceutical compositions provided herein are formulated for oral administration in tablet, capsule, powder, or liquid form.
  • a tablet comprises a solid carrier or an adjuvant.
  • Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil, or synthetic oil.
  • physiological saline solution, dextrose or other saccharide solution, or glycols are optionally included.
  • a capsule comprises a solid carrier such as gelatin.
  • the pharmaceutical compositions are provided in a dosage form for parenteral administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers.
  • a parenterally acceptable aqueous solution which is pyrogen-free and has a suitable pH, isotonicity, and stability.
  • isotonic vehicles such as Sodium Chloride injection, Ringer’s injection, or Lactated Ringer’s injection.
  • preservatives, stabilizers, buffers, antioxidants, and/or other additives are included.
  • compositions are provided in a dosage form for topical administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers.
  • composition comprising at least one heterocyclic RBP4 inhibitory compound as described herein differ, depending upon the patient's condition, that is, stage of the disease, general health status, age, and other factors.
  • compositions are administered in a manner appropriate to the disease to be treated (or prevented).
  • An appropriate dose and a suitable duration and frequency of administration will be determined by such factors as the condition of the patient, the type and severity of the patient's disease, the particular form of the active ingredient, and the method of administration.
  • an appropriate dose and treatment regimen provides the composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (e.g., an improved clinical outcome), or a lessening of symptom severity.
  • Optimal doses are generally determined using experimental models and/or clinical trials. The optimal dose depends upon the body mass, weight, or blood volume of the patient.
  • the compounds described herein, or a pharmaceutically acceptable salt thereof are used in the preparation of medicaments for the treatment of diseases or conditions in a mammal that would benefit from administration of any one of the compounds disclosed.
  • Methods for treating any of the diseases or conditions described herein in a mammal in need of such treatment involves administration of pharmaceutical compositions that include at least one compound described herein or a pharmaceutically acceptable salt, active metabolite, prodrug, or
  • compositions containing the compound(s) described herein are administered for prophylactic and/or therapeutic treatments.
  • the compositions are administered to a patient already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest at least one of the symptoms of the disease or condition. Amounts effective for this use depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and the judgment of the treating physician.
  • Therapeutically effective amounts are optionally determined by methods including, but not limited to, a dose escalation and/or dose ranging clinical trial.
  • compositions containing the compounds described herein are administered to a patient susceptible to or otherwise at risk of a particular disease, disorder or condition. Such an amount is defined to be a“prophylactically effective amount or dose.”
  • a“prophylactically effective amount or dose” In this use, the precise amounts also depend on the patient's state of health, weight, and the like. When used in patients, effective amounts for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician.
  • prophylactic treatments include administering to a mammal, in which the mammal previously experienced at least one symptom of the disease being treated and is currently in remission, a pharmaceutical composition comprising a compound described herein, or a pharmaceutically acceptable salt thereof, in order to prevent a return of the symptoms of the disease or condition.
  • the administration of the compounds are administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition.
  • Oral doses typically range from about 1.0 mg to about 1000 mg, one to four times, or more, per day. In general, however, doses employed for adult human treatment are typically in the range of 0.01 mg to 5000 mg per day. In one aspect, doses employed for adult human treatment are from about 1 mg to about 1000 mg per day. In one embodiment, the desired dose is conveniently presented in a single dose or in divided doses administered simultaneously or at appropriate intervals, for example as two, three, four or more sub-doses per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, about 100 mg, about 105 mg, about 110 mg, about 115 mg, about 120 mg, about 125 mg, about 130 mg, about 135 mg, about 140 mg, about 145 mg, about 150 mg, about 155 mg, about 160 mg, about 165 mg, about 170 mg, about 175 mg, about 180 mg, about 185 mg, about 190 mg, about 195 mg, about 200 mg, about 225 mg, about 240 mg, about 250
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to about 15 mg, up to about 20 mg, up to about 25 mg, up to about 30 mg, up to about 35 mg, up to about 40 mg, up to about 45 mg, up to about 50 mg, up to about 55 mg, up to about 60 mg, up to about 65 mg, up to about 70 mg, up to about 75 mg, up to about 80 mg, up to about 85 mg, up to about 90 mg, up to about 95 mg, up to about 100 mg, up to about 105 mg, up to about 110 mg, up to about 115 mg, up to about 120 mg, up to about 125 mg, up to about 130 mg, up to about 135 mg, up to about 140 mg, up to about 145 mg, up to about 150 mg, up to about 155 mg, up to
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 35 mg, at least 40 mg, at least 45 mg, at least 50 mg, at least 55 mg, at least 60 mg, at least 65 mg, at least 70 mg, at least 75 mg, at least 80 mg, at least 85 mg, at least 90 mg, at least 95 mg, at least 100 mg, at least 105 mg, at least 110 mg, at least 115 mg, at least 120 mg, at least 125 mg, at least 130 mg, at least 135 mg, at least 140 mg, at least 145 mg, at least 150 mg, at least 155 mg, at least 160 mg, at least 165 mg, at least 170 mg, at least 175 mg, at least 180 mg, at least
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 25 mg, about 50 mg, about 75 mg, about 100 mg, about 150 mg, about 200 mg, or about 400 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 50 mg, about 100 mg, about 150 mg, about 200 mg, or about 400 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 25 mg, up to 50 mg, up to 75 mg, up to 100 mg, up to 150 mg, up to 200 mg, or up to 400 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least 25 mg, at least 50 mg, at least 75 mg, at least 100 mg, at least 150 mg, at least 200 mg, or at least 400 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 25 mg, up to 50 mg, up to 75 mg, up to 100 mg, up to 150 mg, up to 200 mg, up to 400 mg, up to 600 mg, up to 800 mg, or up to 1000 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 25 mg per day, about 50 mg per day, about 75 mg per day, about 100 mg per day, about 150 mg per day, about 200 mg per day, or about 400 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 50 mg per day, about 100 mg per day, about 150 mg per day, about 200 mg per day, or about 400 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 25 mg per day, up to 50 mg per day, up to 75 mg per day, up to 100 mg per day, up to 150 mg per day, up to 200 mg per day, or up to 400 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least 25 mg per day, at least 50 mg per day, at least 75 mg per day, at least 100 mg per day, at least 150 mg per day, at least 200 mg per day, or at least 400 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 25 mg per day, up to 50 mg per day, up to 75 mg per day, up to 100 mg per day, up to 150 mg per day, up to 200 mg per day, up to 400 mg per day, up to 600 mg per day, up to 800 mg per day, or up to 1000 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 400 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 200 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 150 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 100 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 75 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 50 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 25 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 400 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 200 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 150 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 100 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 75 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 50 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of up to 25 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 400 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 200 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 150 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 100 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 75 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 50 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 25 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 400 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 200 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 150 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 100 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 75 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 50 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of about 25 mg per day.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 400 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 200 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 150 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 100 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 75 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 50 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 25 mg.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 400 mg per day.
  • a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 200 mg per day.
  • a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 150 mg per day.
  • a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 100 mg per day.
  • a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 75 mg per day.
  • a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 50 mg per day.
  • pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered to a subject or patient in an amount of at least about 25 mg per day.
  • the daily dosages appropriate for the compound described herein, or a pharmaceutically acceptable salt thereof are from about 0.01 to about 50 mg/kg per body weight.
  • the daily dosage or the amount of active in the dosage form are lower or higher than the ranges indicated herein, based on a number of variables in regard to an individual treatment regime.
  • the daily and unit dosages are altered depending on a number of variables including, but not limited to, the activity of the compound used, the disease or condition to be treated, the mode of administration, the requirements of the individual subject, the severity of the disease or condition being treated, and the judgment of the practitioner.
  • a maintenance dose is administered if necessary. Subsequently, in specific embodiments, the dosage or the frequency of administration, or both, is reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained. In certain embodiments, however, the patient requires intermittent treatment on a long-term basis upon any recurrence of symptoms.
  • Toxicity and therapeutic efficacy of such therapeutic regimens are determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 and the ED50.
  • the dose ratio between the toxic and therapeutic effects is the therapeutic index and it is expressed as the ratio between LD50 and ED50.
  • the data obtained from cell culture assays and animal studies are used in formulating the therapeutically effective daily dosage range and/or the therapeutically effective unit dosage amount for use in mammals, including humans.
  • the daily dosage amount of the compounds described herein lies within a range of circulating concentrations that include the ED50 with minimal toxicity.
  • the daily dosage range and/or the unit dosage amount varies within this range depending upon the dosage form employed and the route of administration utilized.
  • the effective amount of the compound described herein, or a pharmaceutically acceptable salt thereof is: (a) systemically administered to the mammal; and/or (b) administered orally to the mammal; and/or (c) intravenously administered to the mammal; and/or (d) administered by injection to the mammal; and/or (e) administered topically to the mammal; and/or (f) administered non-systemically or locally to the mammal.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered orally or parenterally to the subject in need thereof.
  • Parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, intravesical, and subcutaneous administration.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered orally or intravenously to a subject in need thereof.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N- oxide, stereoisomer, or isomer thereof is administered orally to a subject in need thereof.
  • a compound of Formula (I), Formula (II), or Formula (III), or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof is administered intravenously to a subject in need thereof.
  • administrations of the effective amount of the compound including further embodiments in which (i) the compound is administered once a day; or (ii) the compound is administered to the mammal multiple times over the span of one day, e.g., two, three, four or more times daily.
  • the heterocyclic RBP4 inhibitory compounds described herein are administered daily, every other day, every other day 3 times a week, every 2 weeks, every 3 weeks, every 4 weeks, every 5 weeks, every 3 days, every 4 days, every 5 days, every 6 days, weekly, bi-weekly, 3 times a week, 4 times a week, 5 times a week, 6 times a week, once a month, twice a month, 3 times a month, once every 2 months, once every 3 months, once every 4 months, once every 5 months, or once every 6 months.
  • the heterocyclic RBP4 inhibitory compounds described herein, or a pharmaceutically acceptable salt, solvate, polymorph, prodrug, metabolite, N-oxide, stereoisomer, or isomer thereof are administered daily.
  • any of the aforementioned aspects are further embodiments comprising multiple administrations of the effective amount of the compound, including further embodiments in which (i) the compound is administered continuously or intermittently: as in a single dose; (ii) the time between multiple administrations is every 6 hours; (iii) the compound is administered to the mammal every 8 hours; (iv) the compound is administered to the mammal every 12 hours; (v) the compound is administered to the mammal every 24 hours.
  • the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (e.g., a “drug holiday”).
  • the length of the drug holiday is between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, or more than 28 days.
  • the dose reduction during a drug holiday is, by way of example only, by 10%-100%, including by way of example only 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, and 100%.
  • the method comprises a drug holiday, wherein the administration of the compound is temporarily suspended or the dose of the compound being administered is temporarily reduced; at the end of the drug holiday, dosing of the compound is resumed.
  • the length of the drug holiday varies from 2 days to 7 days. In one embodiment, the length of the drug holiday is 7 days. In one embodiment, the length of the drug holiday is 14 days. In one embodiment, the length of the drug holiday is 28 days.
  • Example 1 Compound 1 Single Dose Non-Human Primate PK Study
  • Example 2 Compound 1 Single Dose Rat PK Study
  • Example 3 Compound 1 Repeat Dose 7-Day Rat PK Study
  • Example 4 Compound 1 Single Dose Mouse PK Study
  • Example 5 Compound 1 Single Dose Dog PK Study
  • Subjects The subject population consists of healthy volunteers.
  • Objects of the study are to characterize the systemic and hepatic safety of single and multiple doses of Compound 1 or Compound 2, characterize the pharmacokinetics (PK) of single and multiple doses of Compound 1 or Compound 2, and determine effects of single and multiple doses of Compound 1 or Compound 2 on serum levels of retinol binding protein 4 (RBP4), a pharmacodynamics (PD) marker.
  • PK pharmacokinetics
  • RBP4 retinol binding protein 4
  • PD pharmacodynamics
  • Study Design and Duration The study is a randomized, double blind, placebo-controlled, sequential single and multiple ascending dose study. There are 5 dose levels in the single and 5 dose levels in the multiple ascending dose components. Each cohort consists of 8 subjects, 6 receiving compound 1 or compound 2 and 2 receiving placebo. Consistent with NINDS policies, 3-5 subjects in each cohort are female and efforts should be made to include diverse races and ethnicity. All subjects participate in a screening period lasting up to 28 days, during which they are assessed for eligibility.
  • Cohorts 1-3 reside in the research unit from Day -1 through Day 3; Cohorts 4 and 5 on Days -1 to 4. On Day -1 baseline safety and PD markers are obtained. Subjects receive a single oral dose on the morning of Day 1. Safety, PK and PD are obtained through Day 3 in early cohorts, though Day 4 in later. A phone call to ask about adverse events and concomitant medications takes place on Day 6. Subjects return for an outpatient visit on Day 8.
  • Cohorts 1-3 reside in the research unit from Day -1 to Day 3; Cohorts 4 and 5 from Day -1 to Day 4. Subjects receive the first oral dose on the morning of Day 1. Safety, PK and PD are obtained through Day 3 or 4, as appropriate, and subjects are discharged from the research unit with investigational product (IP) to take as an outpatient. Subjects return for outpatient visits on Days 6, 8, and 12. They are readmitted to the unit on the morning of Day 15 and receive the final oral dose in the research unit. Safety, PK and PD are collected until discharge on Day 17 for Cohorts 1-3, day 18 for Cohorts 4 and 5. Subjects return for a last safety visit on Day 22. Specifics regarding safety, PK and PD collection for both SAD and MAD are described verbally in following sections, and presented in tabular form in Tables 5-10.
  • Safety evaluation is evaluated by collecting adverse events, physical examination, Ocular examination (including slit lamp biomicroscopy, dilated ophthalmoscopy and intraocular pressure), Visual acuity, D-28 color vision text, Visual fields, Night vision questionnaire (multiple dose only), Vital signs (blood pressure, heart rate, temperature, respiratory rate), Weight, CBC with differential and platelets, serum chemistry, urinalysis, and ECG.
  • ECGs Twelve lead ECG are performed following recommendations in ICH E14 regarding evaluation of QTc prolongation in patients in early stage clinical trials. ECGs are performed in triplicate on Days 1, 2, 8, 15 and 16. Visual acuity is measured using Early Treatment of Diabetic Retinopathy Study (ETDRS) Visual Acuity charts 1 and 2. The chart is placed on an ETDRS light box which is hung at eye level on the wall or placed on a stand. Room lighting is at office levels and uniform between the subject and the light box. The distance from the patient’s eyes to the Visual Acuity Chart is 4.0 meters. If vision tests are performed on the same day as an ERG, they are completed prior to pupil dilation.
  • EDRS Early Treatment of Diabetic Retinopathy Study
  • PK sampling of plasma is conducted in all subjects (for example predose, 0.5, 1, 1.5, 2, 3, 4, 8, 8, 10, 12, 16, 24, 36 and 48 hours post dose on Days 1-3 and 15-17; trough predose on other days; and times are finalized based on toxicokinetics).
  • Compound 1 and compound 2 concentrations are determined using a high-pressure liquid chromatography coupled with a mass spectrometer (LC/MS/MS) method. The method is determined during toxicokinetic studies and then validated for human application. Validation includes determination of the limits of quantitation. If toxicokinetics show significant urinary excretion, then urine PK methods would be added. If toxicokinetics show significant metabolites, this method would be added.
  • ISCEV International Society for Clinical Electrocardiography of Vision
  • Statistical Analysis All subjects who receive IP are included in the safety population. All subjects who receive IP and have at least one post-treatment sample or determination are included in the PK analysis population. Analysis is by treatment assignment. Safety is evaluated by monitoring AEs, and by change from baseline on examinations and laboratory studies. The AEs are coded using the Medical Dictionary for Regulatory Activities (MedDRA) and summarized by system organ class (SOC) and preferred term, by severity, by relationship to study drug and study procedure, and by study drug dose. ECG parameters analyzed include heart rate, PR, QRS, QT, QTcB and QTcF intervals. Analysis of other examinations are specified in the protocol.
  • MedDRA Medical Dictionary for Regulatory Activities
  • SOC system organ class
  • ECG parameters analyzed include heart rate, PR, QRS, QT, QTcB and QTcF intervals. Analysis of other examinations are specified in the protocol.
  • NASH nonalcoholic steatohepatitis
  • Subjects with NAFLD or NASH are treated with doses of either 50 or 100 mg/day of compound 1 or compound 2 for 6 months.
  • Subjects are permitted their usual other medications (e.g. antidiabetic medications such as metformin or sulfonamides) but not glitazones, PPAR agonists, OCA, or similar medications.
  • the subjects are assessed before the study, and at intervals during the study, such as every 4 weeks during the study and 4 weeks after the last dose of compound 1 or compound 2, for safety and pharmacodynamic evaluations.
  • MRIs of the subjects' livers are taken every 4 weeks during the study and 4 weeks after completion of compound 1 or compound 2 dosing, to determine hepatic fat; and liver biopsies are taken before the study (to establish the diagnosis) and 4 weeks after completion of compound 1 or compound 2 dosing.
  • blood is drawn and urine collected; and a standard metabolic panel, complete blood count, and standard urinalysis are performed. Blood is analyzed for total cholesterol, HDL-C, LDL-C, VLDL-C, TGs, apoB, and liver transaminases.
  • the subjects also maintain health diaries, which are reviewed at each visit.
  • the subjects show a dose- related improvement in their disease, as manifested by, for example, MRI and liver biopsy.
  • Overweight subjects are treated with doses of either 50 or 100 mg/day of compound 1 or compound 2 for 2 months.
  • the subjects are not coffee drinkers or consumers of any other form of caffeine.
  • the subjects are not to alter any other behavioral parameter such as diet or exercise.
  • the subjects are assessed before the study, at intervals during the study, such as every 2 weeks during the study and 4 weeks after the last dose of compound 1 or compound 2, for safety and
  • Subjects diagnosed with type 2 diabetes mellitus are treated with doses of either 50 or 100 mg/day of compound 1 or compound 2 for 4 months.
  • the subjects are not to alter any other behavioral parameter such as diet or exercise.
  • the subjects are assessed before the study, at intervals during the study, such as every 2 weeks during the study and 4 weeks after the last dose of compound 1 or compound 2, for safety and pharmacodynamic evaluations, including markers of blood glucose, interstitial glucose, and insulin.
  • the fluoroangiography and retinography are carried out according to the standard methods and the photograms are graduated by giving a score to the plasmatic exudation and to the hard exudates on the basis of the ERDRS Classification.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des composés dérivés hétérocycliques et des compositions pharmaceutiques comprenant lesdits composés qui sont utiles pour le traitement de maladies liées à la protéine de liaison du rétinol (RBP4), telles que l'obésité et autres.
PCT/US2018/037602 2017-06-15 2018-06-14 Méthodes de traitement de maladies métaboliques avec des pyrroles bicycliques fusionnés WO2018232158A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762520381P 2017-06-15 2017-06-15
US62/520,381 2017-06-15

Publications (1)

Publication Number Publication Date
WO2018232158A1 true WO2018232158A1 (fr) 2018-12-20

Family

ID=64658688

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/037602 WO2018232158A1 (fr) 2017-06-15 2018-06-14 Méthodes de traitement de maladies métaboliques avec des pyrroles bicycliques fusionnés

Country Status (1)

Country Link
WO (1) WO2018232158A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007049224A1 (fr) * 2005-10-25 2007-05-03 Actelion Pharmaceuticals Ltd Nouveaux derives d'hexahydro- ou d'octahydro-cyclopenta[c]pyrrole
US20120010186A1 (en) * 2009-03-23 2012-01-12 Merck Frosst Canada Ltd. Heterocyclic compounds as inhibitors of stearoyl-coenzyme a delta-9 desaturase
US20150225342A1 (en) * 2013-09-26 2015-08-13 Mnemosyne Pharmaceuticals, Inc. Selective octahydro-cyclopenta[c] pyrrole negative modulators of nr2b

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007049224A1 (fr) * 2005-10-25 2007-05-03 Actelion Pharmaceuticals Ltd Nouveaux derives d'hexahydro- ou d'octahydro-cyclopenta[c]pyrrole
US20120010186A1 (en) * 2009-03-23 2012-01-12 Merck Frosst Canada Ltd. Heterocyclic compounds as inhibitors of stearoyl-coenzyme a delta-9 desaturase
US20150225342A1 (en) * 2013-09-26 2015-08-13 Mnemosyne Pharmaceuticals, Inc. Selective octahydro-cyclopenta[c] pyrrole negative modulators of nr2b

Similar Documents

Publication Publication Date Title
US20210228563A1 (en) Methods of treating rbp4 related diseases with triazolopyridines
EP1904439B1 (fr) Inhibiteurs de la sphingosine kinase
EP2070926A1 (fr) Dérivés de 13,13a-dihydroberbérine, leur composition pharmaceutique et leurs utilisations
JP2021526123A (ja) オートタキシン阻害剤とその使用
US11389444B2 (en) Methods of treating metabolic diseases with fused bicyclic pyrazoles
KR20060079122A (ko) 당뇨 및 비만 치료예방에 유효한 벤조티아졸 유도체
US10487058B2 (en) Cyclopropyl unsaturated quinoline compound used as leukotriene receptor antagonist and applications thereof
CN115385899B (zh) ROCK2抑制剂belumosudil的氘代药物及用途
WO2018232158A1 (fr) Méthodes de traitement de maladies métaboliques avec des pyrroles bicycliques fusionnés
TW202003450A (zh) 脂肪酸類似物及使用方法
TWI841532B (zh) 以稠合雙環吡唑治療代謝疾病之方法
EP2405751B1 (fr) Pro-médicaments inhibiteurs de la sphingosine kinase
WO2018232160A1 (fr) Méthodes de traitement de maladies métaboliques avec des phénoxyéthylamides
JP4104986B2 (ja) 前立腺肥大症に伴う膀胱刺激症状治療剤
CN101903352A (zh) 取代的3-羟基吡啶系化合物及其药物组合物
US20230357148A1 (en) Indole derivative and application thereof
KR102465988B1 (ko) 항암, 항염증 효능을 가진 삼원환의 약학적 조성물
CN111285843B (zh) 新型5-羟色胺和去甲肾上腺素双重再摄取抑制剂及其医药用途
CN111196816B (zh) 一种七元环化合物及在防治糖尿病及代谢综合征中的应用
CN101735218A (zh) 哌啶氨基甲酸酯衍生物及其应用
CS221846B2 (en) Method of making the ethanolamine derivatives
CN114874202A (zh) 一种用于治疗阿尔茨海默症的药物及其用途
CN116554127A (zh) 哌嗪取代苯酚类衍生物及其用途
JP2023513922A (ja) 3-アリールオキシ-3-5員ヘテロアリール-プロピルアミン化合物の調製方法および結晶形
KR101048748B1 (ko) 신규 갈바닉산 유도체 또는 이의 약학적으로 허용가능한 염, 이의 제조방법 및 이를 유효성분으로 함유하는 다약제내성 억제용 약학적 조성물

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: 18818913

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: 18818913

Country of ref document: EP

Kind code of ref document: A1