WO2019139919A1 - Acetal compounds and therapeutic uses thereof - Google Patents

Acetal compounds and therapeutic uses thereof Download PDF

Info

Publication number
WO2019139919A1
WO2019139919A1 PCT/US2019/012762 US2019012762W WO2019139919A1 WO 2019139919 A1 WO2019139919 A1 WO 2019139919A1 US 2019012762 W US2019012762 W US 2019012762W WO 2019139919 A1 WO2019139919 A1 WO 2019139919A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
optionally substituted
group
ealkyl
ealkoxy
Prior art date
Application number
PCT/US2019/012762
Other languages
French (fr)
Inventor
Lin Zhi
Original Assignee
Ligand Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ligand Pharmaceuticals, Inc. filed Critical Ligand Pharmaceuticals, Inc.
Priority to KR1020207022964A priority Critical patent/KR20200123106A/en
Priority to CN201980016007.3A priority patent/CN111788196A/en
Priority to RU2020126177A priority patent/RU2020126177A/en
Priority to US16/960,681 priority patent/US11970482B2/en
Priority to CA3087932A priority patent/CA3087932A1/en
Priority to EP19738393.8A priority patent/EP3737676B1/en
Priority to JP2020537621A priority patent/JP2021509907A/en
Priority to AU2019207625A priority patent/AU2019207625A1/en
Publication of WO2019139919A1 publication Critical patent/WO2019139919A1/en

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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero 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
    • A61K31/397Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having four-membered rings, e.g. azetidine

Definitions

  • the present disclosure relates to the field of chemistry and medicine. More specifically, the present disclosure relates to acetal and cyclic acetal compounds, compositions, their preparation, and their use as therapeutic agents.
  • Statins are a class of liver-targeting compounds that inhibit HMG-CoA reductase, one of the liver enzymes in the cholesterol biosynthetic pathway. Statins are used as lipid-lowing medicines for the prophylactic and therapeutic treatment of coronary heart disease. Despite their ubiquitous use, statins, like other liver-targeting drugs, suffer from poor oral bioavailability and inadequate distribution to the liver. In fact, a significant portion of patients cannot take statins because of side effects from statin biological activity outside of the liver.
  • liver-targeting compounds and strategies to increase the bioavailability of liver-targeting drugs, to increase drug distribution to the liver, and to reduce drug biological activity outside of the liver.
  • R 1 , R 2 , R 3 , and R 4 have any of the values described herein.
  • R 1 is selected from the group consisting of optionally substituted Ci-galkyl, -COOR 5 , optionally substituted C2-ioalkoxyalkyl, and optionally substituted 3-12 membered heterocyclyl.
  • R 1 and R 2 together with the atoms to which they are attached form an optionally substituted 4-10 membered heterocyclyl, or R 1 and R 3 together with the atoms to which they are attached form an optionally substituted 6-10 membered heterocyclyl.
  • R 2 in some embodiments is selected from the group consisting of H, - C(0)R 6 , and optionally substituted C2-ioalkoxyalkyl.
  • R 1 and R 2 together with the atoms to which they are attached form an optionally substituted 4-10 membered heterocyclyl, or R 2 and R 3 together with the atoms to which they are attached form a 6-10 membered heterocyclyl, optionally substituted with one or more R 7 .
  • R 3 is selected from the group consisting of H, - C(0)R 8 , and optionally substituted Ca-ioalkoxyalkyl.
  • R 1 and R 3 together with the atoms to which they are attached form an optionally substituted 6-10 membered heterocyclyl, or R 2 and R 3 together with the atoms to which they are attached form a 6-10 membered heterocyclyl, optionally substituted with one or more R 7 .
  • R 4 in some embodiments is a fragment of a therapeutic agent.
  • each of R 5 , R 6 , and R 8 is independently selected from the group consisting of H, optionally substituted Ci-ealkyl, halogen, optionally substituted Ca ⁇ alkenyl, optionally substituted C 2 -6alkenyl, optionally substituted C3- 7carbocyclyl, optionally substituted C 6 -ioaryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl.
  • each R 7 is independently selected from the group consisting of optionally substituted Ci-ealkyl, oxo, optionally substituted C 6 -isaryl, and optionally substituted 5-18 membered heteroaryl. [0012] In some embodiments, at least one of R 2 and R 3 is not H, or R 1 is not -
  • Z is O, S, or NR 40 .
  • R 37 in some embodiments is selected from the group consisting of H, a fragment of a therapeutic agent, optionally substituted C1-8alkyl, Ci-6haloalkyl, Ci- eheteroalkyl, optionally substituted Ca-vcarbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C 6 -isaiyl, 5-10 membered heteroaryl, and optionally substituted Ca-ioalkoxy alkyl.
  • each of R 38 and R 39 is independently selected from the group consisting of a fragment of a therapeutic agent, optionally substituted Ci-ealkyl, Ci- ehaloalkyl, C1-6 heteroalkyl, optionally substituted C3-7carbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C1-6 saryl, optionally substituted C2- loalkoxyalkyl, and optionally substituted 5-10 membered heteroaryl.
  • At least one of R 37 , R 38 , and R 39 is a fragment of a therapeutic agent.
  • the fragment of a therapeutic agent is further optionally substituted with one or more moieties selected from the group consisting of i-ealkyl, halo, and C 6 -isaryl.
  • each A is independently selected from O, S, and NR 43 , provided that at least one A is O.
  • each of R 41 , R 42 , and R 43 is independently selected from the group consisting of H, optionally substituted Ci ⁇ alkyl, Ci-ehaloalkyl, Ci- eheteroalkyl, optionally substituted C3-7carbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C 6 -isaryl, optionally substituted Cz-ioalkoxy alkyl, and optionally substituted 5-10 membered heteroaryl.
  • R 40 is independently selected from the group consisting of optionally substituted Ci-ealkyl, Ci-ehaloalkyl, Ci-eheteroalkyl, optionally substituted C3-7carbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C 6 -isaryl, optionally substituted C 2 -ioalkoxyalkyl, and optionally substituted 5-10 membered heteroaryl.
  • liver-targeting compounds containing an acetal moiety are provided that afford increased bioavailability relative to their active drag counterparts, increased drug distribution to the liver relative to their active drug counterparts, and/or reduced biological activity outside of the liver.
  • Various embodiments of these compounds include compounds having the structures of Formula I or P as described above or pharmaceutically acceptable salts thereof.
  • the compounds of Formula I are also represented by the structure of Formula la, lb, Ic, Id, Ie, If, or Ig:
  • X is an optionally substituted Cl-7alkylene linker.
  • X is a Cl-7alkylene linker optionally substituted with one or more Rl l.
  • Rl l is selected from the group consisting of Cl-6alkyl, Cl-6alkenyl, Cl-6alkynyl, Cl-6heteroalkyl, C3- 7carbocyclyl, 3-10 member ed heterocyclyl, C6-18aryl, C6-18arylCl-6alkyl, 5-10 membered heteroaryl, 5-10 membered heteroarylCl-6alkyl, halo, cyano, hydroxy, Cl-6alkoxy, C2- lOalkoxyalkyl, C6-18aiyloxy, sulfhydryl, haloCl-6alkyl, haloCl-6alkoxy, Cl-6alkylthio, C6-18arylthi
  • Rll is selected from the group consisting of Cl-6alkyl, C3-7carbocyclyl, Cl-6alkoxy, C6-18aryl, or halo.
  • X is a C2alkylene linker. In some particular embodiments, X is -CH2CH2-. In some further embodiments, X is a C3alkylene linker. In some particular embodiments, X is - CH2CH2CH2-. In some particular embodiments, X is -CH2(CH2CH3)CH2-. In other particular embodiments, X is -CH2(CH3)CH2CH2(CH3)-.
  • RIO is independently an optionally substituted Cl-6alkyl.
  • each of R9 and RIO is independently a Cl-6alkyl optionally substituted with one or more R12.
  • R12 is selected from the group consisting of Cl-6alkyl, Cl- 6alkenyl, Cl-6alkynyl, Cl-6heteroalkyl, C3-7carbocyclyl, C3-7carbocyclylCl-6alkyl, 3-10 membered heterocyclyl, 3-10 membered heterocyclylCl-6alkyl, C6-18aryl, C6-18arylCl- 6alkyl, 5-10 membered heteroaryl, 5-10 membered heteroarylCl-6alkyl, and haloCl-6alkyl.
  • R9 is a Cl-6alkyl. In some particular embodiments, R9 is ethyl. In some further embodiments, R10 is a Cl-6alkyl. In some particular embodiments, R10 is ethyl.
  • Y is a Cl-5alkylene linker, optionally substituted with one or more R7.
  • R7 is selected from the group consisting of optionally substituted Cl-8alkyl, oxo, optionally substituted C6-18aryl, and optionally substituted 5-18 membered heteroaryl.
  • Y is a Clalkylene linker.
  • R1 is an optionally substituted Cl-8alkyl.
  • R1 is a Cl-8alkyl optionally substituted with one or more R13.
  • R13 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro.
  • R1 is -CH20H.
  • R1 is - COOR5. In some such embodiments, R5 is H. In other such embodiments, R5 is methyl.
  • R1 is an optionally substituted C2-10alkoxyalkyl. In some such embodiments, R1 is a C2- lOalkoxyalkyl optionally substituted with one or more R14.
  • R14 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro.
  • R1 is -CH20CH20CH2CH3.
  • R1 is - CH(OCH2CH3)2.
  • R1 is an optionally substituted 3-12 membered heterocyclyl. In some such embodiments, R1 is a 3-12 membered heterocyclyl optionally substituted with one or more R15.
  • R15 is selected from the group consisting of Cl-6alkyl, Cl-6alkenyl, Cl- 6alkynyl, Cl-6heteroalkyl, C3-7carbocyclyl, 3-10 membered heterocyclyl, C6-18aryl, C6- 18arylCl-6alkyl, 5-10 membered heteroaryl, 5-10 membered heteroarylCl-6alkyl, halo, cyano, hydroxy, Cl-6alkoxy, C2-10alkoxyalkyl, aryloxy, sulfhydryl, haloCl-6alkyl, haloCl- 6alkoxy, Cl-6alkylthio, C6-18arylthio, and nitro.
  • R1 is a 5
  • R1 is or
  • R1 is a 6 membered heterocyclyl.
  • R1 is
  • R2 is H.
  • R2 is -C(0)R6.
  • R6 is an optionally substituted Cl-6alkyl.
  • R6 is a Cl-6alkyl optionally substituted with one or more R16.
  • R16 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6- lOarylthio, and nitro.
  • R6 is -CH(CH3)2.
  • R2 is an optionally substituted C2-10alkoxyalkyl.
  • R2 is a C2-10alkoxyalkyl optionally substituted with one or more R17.
  • R17 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6-10aryloxy, sulfhydiyl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro.
  • R2 is -CH20CH2CH3.
  • R3 is H.
  • R3 is -C(0)R8.
  • R8 is an optionally substituted Cl-6alkyl.
  • R8 is a Cl-6alkyl optionally substituted with one or more R18.
  • R18 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6- lOarylthio, and nitro.
  • R8 is -CH(CH3)2.
  • R3 is an optionally substituted C2-10alkoxyalkyl.
  • R3 is a C2- lOalkoxy alkyl optionally substituted with one or more R19.
  • R19 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6- lOaryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro.
  • R3 is -CH20CH2CH3.
  • R7 is an optionally substituted Cl-8alkyl.
  • R7 is a Cl-8alkyl optionally substituted with one or more R20.
  • R20 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro.
  • R7 is methyl.
  • R7 is -CH2CH2CH20H.
  • R7 is oxo.
  • R7 is an optionally substituted C6-18aryl. In some such embodiments, R7 is a C6-18aryl optionally substituted with one or more R21.
  • each of R22, R23, R24, R25, R26, R27, and R28 is independently selected from the group consisting of hydrogen, Cl-6alkyl, C2- 6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aiyl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl.
  • R7 is a phenyl.
  • R7 is
  • R7 is an optionally substituted 5-18 membered heteroaryl.
  • R7 is a 5-18 membered heteroaryl optionally substituted with one or more R29.
  • each of R30, R31, R32, R33, R34, R35, and R36 is independently selected from the group consisting of hydrogen, Cl-6alkyl, C2-6alkenyl, C2-6alkynyl, C3- 7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl.
  • R7 is a 6 membered heteroaryl.
  • R4 is a fragment of a liver-targeting therapeutic agent or a fragment of a liver-activated therapeutic agent.
  • R4 is a fragment of a lipid-lowering therapeutic agent or a fragment of a cholesterol-biosynthesis-inhibiting therapeutic agent.
  • R4 is a fragment of a HMG-CoA reductase inhibitor.
  • R4 is a fragment of a statin.
  • R4 is selected from the group consisting of
  • R4 is
  • Z is O.
  • R37 is H.
  • R39 in some embodiments is ethyl.
  • R38 in some embodiments is the optionally substituted fragment of a therapeutic agent.
  • the fragment of a therapeutic agent is
  • the substituent is m R38 has the structure
  • the fragment of the therapeutic agent is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • A is O.
  • R41 is H.
  • R42 is ethyl
  • the compounds of Formula I, la, lb, Ic, Id, Ie, If, Ig, or P as described herein are selected from the group consisting of
  • the pharmaceutically acceptable salts are selected from alkaline metal salts or ammonium salts. In some embodiments, the pharmaceutically acceptable salts are sodium salts, including disodium salts.
  • the compounds disclosed herein may exist as individual enantiomers and diastereomers or as mixtures of such isomers, including racemates. Separation of the individual isomers or selective synthesis of the individual isomers is accomplished by application of various methods which are well known to practitioners in the art Unless otherwise indicated, all such isomers and mixtures thereof are included in the scope of the compounds disclosed herein.
  • compounds disclosed herein may exist in one or more crystalline or amorphous forms. Unless otherwise indicated, all such forms are included in the scope of the compounds disclosed herein, including any polymorphic forms.
  • some of the compounds disclosed herein may form solvates with water (i.e., hydrates) or common organic solvents. Unless otherwise indicated, such solvates are included in the scope of the compounds disclosed herein.
  • Isotopes may be present in the compounds described. Each chemical element as represented in a compound structure may include any isotope of said element.
  • a hydrogen atom may be explicitly disclosed or understood to be present in the compound.
  • the hydrogen atom can be any isotope of hydrogen, including but not limited to hydrogen- 1 (protium) and hydrogen-2 (deuterium).
  • reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise.
  • “C a to Cb” or“C a -b” in which“a” and“b” are integers refer to the number of carbon atoms in the specified group. That is, the group can contain from “a” to“b”, inclusive, carbon atoms.
  • a“Ci to Ct alkyl” or“CM alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH3-, CH3CH2-, CH3CH2CH2-, (CH 3 )2CH-, CH3CH2CH2CH2-, CH3CH2CH(CH3)- and (C3 ⁇ 4) 3 C-.
  • halogen or“halo,” as used herein, means any one of the radiostable atoms of column 7 of the Periodic Table of the Elements, e.g., fluorine, chlorine, bromine, or iodine, with fluorine and chlorine being preferred.
  • alkyl refers to a straight or branched hydrocarbon chain that is fully saturated (i.e., contains no double or triple bonds).
  • the alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as“1 to 20” refers to each integer in the given range; e.g.,“1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term“alkyl” where no numerical range is designated).
  • the alkyl group may also be a medium size alkyl having 1 to 9 carbon atoms.
  • the alkyl group could also be a lower alkyl having 1 to 4 carbon atoms.
  • the alkyl group may be designated as“CM alkyl” or similar designations.
  • “CM alkyl” indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from the group consisting of methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, and the like.
  • alkoxy refers to the formula -OR wherein R is an alkyl as is defined above, such as“C1-9 alkoxy”, including but not limited to methoxy, ethoxy, n- propoxy, l-methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, and tert-butoxy, and the like.
  • alkylthio refers to the formula -SR wherein R is an alkyl as is defined above, such as“C1-9 alkylthio” and the like, including but not limited to methylmercapto, ethylmercapto, n-propylmercapto, 1 -methylethy lmercapto
  • alkenyl refers to a straight or branched hydrocarbon chain containing one or more double bonds.
  • the alkenyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term“alkenyl” where no numerical range is designated.
  • the alkenyl group may also be a medium size alkenyl having 2 to 9 carbon atoms.
  • the alkenyl group could also be a lower alkenyl having 2 to 4 carbon atoms.
  • the alkenyl group may be designated as “C2-* alkenyl” or similar designations.
  • “C 2-4 alkenyl” indicates that there are two to four carbon atoms in the alkenyl chain, i.e., the alkenyl chain is selected from the group consisting of ethenyl, propen-l-yl, propen-2-yl, propen-3-yl, buten-l-yl, buten-2-yl, buten-3-yl, buten- 4-yl, 1 -methyl-propen- 1 -yl, 2-methyl-propen- 1 -yl, 1-ethyl-ethen-l-yl, 2-methyl-propen-3-yl, buta-l,3-dienyl, buta-l,2,-dienyl, and buta-l,2-dien-4-yl.
  • alkenyl groups include, but are in no way limited to, ethenyl, propenyl, butenyl, pentenyl, and hexenyl, and the like.
  • “alkynyl” refers to a straight or branched hydrocarbon chain containing one or more triple bonds.
  • the alkynyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term“alkynyl” where no numerical range is designated.
  • the alkynyl group may also be a medium size alkynyl having 2 to 9 carbon atoms.
  • the alkynyl group could also be a lower alkynyl having 2 to 4 carbon atoms.
  • the alkynyl group may be designated as “C2-4 alkynyl” or similar designations.
  • “C 2-4 alkynyl” indicates that there are two to four carbon atoms in the alkynyl chain, i.e., the alkynyl chain is selected from the group consisting of ethynyl, propyn-l-yl, propyn-2-yl, butyn-l-yl, butyn-3-yl, butyn-4-yl, and 2- butynyl.
  • Typical alkynyl groups include, but are in no way limited to, ethynyl, propynyl, butynyl, pentynyl, and hexynyl, and the like.
  • heteroalkyl refers to a straight or branched hydrocarbon chain containing one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur, in the chain backbone.
  • the heteroalkyl group may have 1 to 20 carbon atom, although the present definition also covers the occurrence of the term“heteroalkyl” where no numerical range is designated.
  • the heteroalkyl group may also be a medium size heteroalkyl having 1 to 9 carbon atoms.
  • the heteroalkyl group could also be a lower heteroalkyl having 1 to 4 carbon atoms.
  • the heteroalkyl group may be designated as“Ci- heteroalkyl” or similar designations.
  • the heteroalkyl group may contain one or more heteroatoms.
  • “CM heteroalkyl” indicates that there are one to four carbon atoms in the heteroalkyl chain and additionally one or more heteroatoms in the backbone of the chain.
  • a heteroalkyl may contain from 1 to 10 heteroatoms, for example, 1 to 5 heteroatoms, 1 to 3 heteroatoms, 2 heteroatoms, or 1 heteroatom.
  • alkylene means a branched, or straight chain fully saturated di-radical chemical group containing only carbon and hydrogen that is attached to the rest of the molecule via two points of attachment (i.e., an alkanediyl).
  • the alkylene group may have 1 to 20 carbon atoms, although the present definition also covers the occurrence of the term alkylene where no numerical range is designated.
  • the alkylene group may also be a medium size alkylene having 1 to 9 carbon atoms.
  • the alkylene group could also be a lower alkylene having 1 to 4 carbon atoms.
  • the alkylene group may be designated as“Ci-4 alkylene” or similar designations.
  • “C1-4 alkylene” indicates that there are one to four carbon atoms in the alkylene chain, i.e., the alkylene chain is selected from the group consisting of methylene, ethylene, ethan- 1,1-diyl, propylene, propan-
  • alkenylene means a straight or branched chain di-radical chemical group containing only carbon and hydrogen and containing at least one carbon- carbon double bond that is attached to the rest of the molecule via two points of attachment.
  • the alkenylene group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term alkenylene where no numerical range is designated.
  • the alkenylene group may also be a medium size alkenylene having 2 to 9 carbon atoms.
  • the alkenylene group could also be a lower alkenylene having 2 to 4 carbon atoms.
  • the alkenylene group may be designated as“C 2-4 alkenylene” or similar designations.
  • “C 2-4 alkenylene” indicates that there are two to four carbon atoms in the alkenylene chain, i.e., the alkenylene chain is selected from the group consisting of ethenylene, ethen-l,l-diyl, propenylene, propen- 1,1-diyl, prop-2-en-l,l-diyl, 1-methyl- ethenylene, but-l-enylene, but-2-enylene, but-l,3-dienylene, buten- 1,1-diyl, but-l,3-dien- 1,1-diyl, but-2-en- 1,1-diyl, but-3-en- 1,1-diyl, l-methyl-prop-2-en-l,l-diyl, 2-methyl-prop-2- en-l,l-diyl, 1-ethyl-ethenylene, 1,2-dimethyl-ethenylene
  • aromatic refers to a ring or ring system having a conjugated pi electron system and includes both carbocyclic aromatic (e.g., phenyl) and heterocyclic aromatic groups (e.g., pyridine).
  • carbocyclic aromatic e.g., phenyl
  • heterocyclic aromatic groups e.g., pyridine
  • the term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of atoms) groups provided that the entire ring system is aromatic.
  • aryl refers to an aromatic ring or ring system (i.e., two or more fused rings that share two adjacent carbon atoms) containing only carbon in the ring backbone. When the aryl is a ring system, every ring in the system is aromatic.
  • the aryl group may have 6 to 18 carbon atoms, although the present definition also covers the occurrence of the term “aryl” where no numerical range is designated. In some embodiments, the aryl group has 6 to 10 carbon atoms.
  • the aryl group may be designated as “C 6 -io aryl,”“C 6 or Cio aryl,” or similar designations. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, azulenyl, and anthracenyl.
  • “aiyloxy” and“arylthio” refers to RO- and RS-, in which R is an aryl as is defined above, such as“C 6 -io aryloxy” or“C 6 -io arylthio” and the like, including but not limited to phenyloxy.
  • An“aralkyl” or“arylalkyl” is an aryl group connected, as a substituent, via an alkylene group, such as“C7-14 aralkyl” and the like, including but not limited to benzyl, 2-phenylethyl, 3-phenylpropyl, and naphthylalkyl.
  • the alkylene group is a lower alkylene group (i.e., a CM alkylene group).
  • heteroaryl refers to an aromatic ring or ring system (i.e., two or more fused rings that share two adjacent atoms) that contain(s) one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur, in the ring backbone.
  • heteroaryl is a ring system, every ring in the system is aromatic.
  • the heteroaryl group may have 5-18 ring members (i.e., the number of atoms making up the ring backbone, including carbon atoms and heteroatoms), although the present definition also covers the occurrence of the term“heteroaryl” where no numerical range is designated.
  • the heteroaryl group has 5 to 10 ring members or 5 to 7 ring members.
  • the heteroaryl group may be designated as “5-7 member ed heteroaryl,”“5-10 membered heteroaryl,” or similar designations.
  • from 1 to 5 are heteroatoms, for example, the heteroaryl may contain from 1 to 4 heteroatoms, from 1 to 3 heteroatoms, two heteroatoms, or one heteroatom.
  • heteroaryl rings include, but are not limited to, furyl, thienyl, phthalazinyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, quinolinyl, isoquinlinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, indolyl, isoindolyl, and benzothienyl.
  • A“heteroaralkyl” or“heteroarylalkyl” is heteroaryl group connected, as a substituent, via an alkylene group. Examples include but are not limited to 2-thienylmethyl, 3-thienylmethyl, furylmethyl, thienylethyl, pyrrolylalkyl, pyridylalkyl, isoxazollylalkyl, and imidazolylalkyl.
  • the alkylene group is a lower alkylene group (i.e., a Ci-4 alkylene group).
  • “carbocyclyl” means a non-aromatic cyclic ring or ring system containing only carbon atoms in the ring system backbone. When the carbocyclyl is a ring system, two or more rings may be joined together in a fused, bridged or spiro- connected fashion. Carbocyclyls may have any degree of saturation provided that at least one ring in a ring system is not aromatic. Thus, carbocyclyls include cycloalkyls, cycloalkenyls, and cycloalkynyls.
  • the carbocyclyl group may have 3 to 20 carbon atoms, although the present definition also covers the occurrence of the term“carbocyclyl” where no numerical range is designated.
  • the carbocyclyl group may also be a medium size carbocyclyl having 3 to 10 carbon atoms.
  • the carbocyclyl group could also be a carbocyclyl having 3 to 6 carbon atoms.
  • the carbocyclyl group may be designated as “C3-6 carbocyclyl” or similar designations.
  • carbocyclyl rings include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,3-dihydro-indene, bicycle[2.2.2]octanyl, adamantyl, and spiro[4.4]nonanyl.
  • A“(carbocyclyljalkyl” is a carbocyclyl group connected, as a substituent, via an alkylene group, such as "C 4-10 (carbocyclyl)alkyl” and the like, including but not limited to, cyclopropylmethyl, cyclobutylmethyl, cyclopropylethyl, cyclopropylbutyl, cyclobutylethyl, cyclopropylisopropyl, cyclopentylmethyl, cyclopentylethyl, cyclohexylmethyl, cyclohexylethyl, cycloheptylmethyl, and the like.
  • the alkylene group is a lower alkylene group.
  • cycloalkyl means a fully saturated carbocyclyl ring or ring system. Examples include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • cycloalkenyl means a carbocyclyl ring or ring system having at least one double bond, wherein no ring in the ring system is aromatic.
  • An example is cyclohexenyl.
  • heterocyclyl means a non-aromatic cyclic ring or ring system containing at least one heteroatom in the ring backbone. Heterocyclyls may be joined together in a fused, bridged or spiro-connected fashion. Heterocyclyls may have any degree of saturation provided that at least one ring in the ring system is not aromatic. The heteroatom(s) may be present in either a non-aromatic or aromatic ring in the ring system.
  • the heterocyclyl group may have 3 to 20 ring members (i.e., the number of atoms making up the ring backbone, including carbon atoms and heteroatoms), although the present definition also covers the occurrence of the term “heterocyclyl” where no numerical range is designated.
  • the heterocyclyl group may also be a medium size heterocyclyl having 3 to 10 ring members.
  • the heterocyclyl group could also be a heterocyclyl having 3 to 6 ring members.
  • the heterocyclyl group may be designated as“3-6 membered heterocyclyl” or similar designations.
  • the heterocyclyl may contain from 1 to 4 heteroatoms, from 1 to 3 heteroatoms, two heteroatoms, or one heteroatom.
  • the heteroatom(s) are selected from one up to three of O, N or S, and in preferred five membered monocyclic heterocyclyls, the heteroatom(s) are selected from one or two heteroatoms selected from O, N, or S.
  • heterocyclyl rings include, but are not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, imidazolidinyl, morpholinyl, oxiranyl, oxepanyl, thiepanyl, piperidinyl, piperazinyl, dioxopiperazinyl, pyrrolidinyl, pyrrolidonyl, pyrrolidionyl, 4-piperidonyl, pyrazolinyl, pyrazolidinyl, 1,3-dioxinyl, 1,3-dioxanyl, 1,4-dioxinyl, 1,4-dioxanyl, 1,3-oxathianyl, 1,4- oxathiinyl, 1,4-oxathianyl, 2H--,2-oxazinyl, trioxanyl, hexan
  • a “(heterocyclyl)alkyl” is a heterocyclyl group connected, as a substituent, via an alkylene group. Examples include, but are not limited to, imidazolinylmethyl and indolinylethyl.
  • Non-limiting examples include formyl, acetyl, propanoyl, benzoyl, and acryl.
  • A“cyano” group refers to a“-CN” group.
  • A“cyanato” group refers to an“-OCN” group.
  • An“isocyanato” group refers to a“-NCO” group.
  • A“thiocyanato” group refers to a“-SCN” group.
  • An“isothiocyanato” group refers to an“-NCS” group.
  • A“sulfonyl” group refers to an“-SO2R” group in which R is selected from hydrogen, optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C 6 -io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
  • An“S-sulfonamido” group refers to a“-SC>2NRARB” group in which RA and RB are each independently selected from hydrogen, halogen, optionally substituted Ci-6 alkyl, optionally substituted Ci-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C 6 -io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
  • An“N-sulfonamido” group refers to a“-N(RA)SC>2RB" group in which RA and RB are each independently selected from hydrogen, halogen, optionally substituted Ci-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C 6 -io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
  • An“amino” group refers to a“-NRARB” group in which RA and RB are each independently selected from hydrogen, halogen, optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C 6 -io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl as defined herein.
  • a nonlimiting example includes free amino (i.e., -NH2).
  • aminoalkyl refers to an amino group connected via an alkylene group.
  • An“alkoxyalkyl” group refers to an alkoxy group connected via an alkylene group, such as a“C2-8 alkoxyalkyl” and the like.
  • a substituted group is derived from the unsubstituted parent group in which there has been an exchange of one or more hydrogen atoms for another atom or group.
  • substituents independently selected from C 1 -C 6 alkyl, C 1 -C 6 alkenyl, C 1 -C 6 alkynyl, C 1 -C 6 heteroalkyl, C3-C7 carbocyclyl (optionally substituted with halo, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, and C 1 -C 6 haloalkoxy), C3- C7-carbocyclyl-Ci-C6-alkyl (optionally substituted with halo, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, Ci-C
  • radical naming conventions can include either a mono-radical or a di-radical, depending on the context. For example, where a substituent requires two points of attachment to the rest of the molecule, it is understood that the substituent is a di-radical.
  • a substituent identified as alkyl that requires two points of attachment includes di-radicals such as -CH2-, -CH2CH2-, -CH 2 CH(CH3)CH2-, and the like.
  • Other radical naming conventions clearly indicate that the radical is a di-radical such as“alkylene” or“alkenylene.”
  • R groups are said to form a ring (e.g., a carbocyclyl, heterocyclyl, aryl, or heteroaryl ring)“together with the atom to which they are attached,” it is meant that the collective unit of the atom and the two R groups are the recited ring.
  • the ring is not otherwise limited by the definition of each R group when taken individually. For example, when the following substructure is present:
  • R 1 and R 2 are defined as selected from the group consisting of hydrogen and alkyl, or R 1 and R 2 together with the nitrogen to which they are attached form a heterocyclyl, it is meant that R 1 and R 2 can be selected from hydrogen or alkyl, or alternatively, the substructure has structure:
  • ring A is a heterocyclyl ring containing the depicted nitrogen.
  • two“adjacent” R groups are said to form a ring“together with the atom to which they are attached,” it is meant that the collective unit of the atoms, intervening bonds, and the two R groups are the recited ring.
  • the following substructure is present:
  • R 1 and R 2 are defined as selected from the group consisting of hydrogen and alkyl, or R 1 and R 2 together with the atoms to which they are attached form an aryl or carbocyclyl, it is meant that R 1 and R 2 can be selected from hydrogen or alkyl, or alternatively, the substructure has structure:
  • A is an aryl ring or a carbocyclyl containing the depicted double bond.
  • a substituent is depicted as a di-radical (i.e., has two points of attachment to the rest of the molecule), it is to be understood that the substituent can be attached in any directional configuration unless otherwise indicated.
  • Subject as used herein, means a human or a non-human mammal, e.g., a dog, a cat, a mouse, a rat, a cow, a sheep, a pig, a goat, a non-human primate or a bird, e.g., a chicken, as well as any other vertebrate or invertebrate.
  • the term“mammal” is used in its usual biological sense. Thus, it specifically includes, but is not limited to, primates, including simians (chimpanzees, apes, monkeys) and humans, cattle, horses, sheep, goats, swine, rabbits, dogs, cats, rodents, rats, mice guinea pigs, or the like.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art.
  • a therapeutic effect relieves, to some extent, one or more of the symptoms of a disease or condition, and includes curing a disease or condition. “Curing” means that the symptoms of a disease or condition are eliminated; however, certain long-term or permanent effects may exist even after a cure is obtained (such as extensive tissue damage).
  • Treatment refers to administering a compound or pharmaceutical composition to a subject for prophylactic and/or therapeutic purposes.
  • prophylactic treatment refers to treating a subject who does not yet exhibit symptoms of a disease or condition, but who is susceptible to, or otherwise at risk of, a particular disease or condition, whereby the treatment reduces the likelihood that the patient will develop the disease or condition.
  • therapeutic treatment refers to administering treatment to a subject already suffering from a disease or condition.
  • Methodabolites of the compounds disclosed herein include active species that are produced upon introduction of the compounds into the biological milieu.
  • Solvate refers to the compound formed by the interaction of a solvent and a compound described herein, a metabolite, or salt thereof. Suitable solvates are pharmaceutically acceptable solvates including hydrates.
  • pharmaceutically acceptable salt refers to salts that retain the biological effectiveness and properties of a compound, which are not biologically or otherwise undesirable for use in a pharmaceutical.
  • the compounds herein are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic 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.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like; particularly preferred are the ammonium, potassium, sodium, calcium and magnesium salts.
  • Organic bases from which salts can be derived include, for example, primaiy, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. Many such salts are known in the art, as described in WO 87/05297, Johnston et al., published September 11, 1987 (incorporated by reference herein in its entirety).
  • the compounds disclosed herein may be synthesized by methods described below, or by modification of these methods. Ways of modifying the methodology include, among others, temperature, solvent, reagents, etc., known to those skilled in the art. In general, during any of the processes for preparation of the compounds disclosed herein, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry (ed. J.F.W. McOmie, Plenum Press, 1973); and P.G.M Green, T.W.
  • Scheme I is provided for the guidance of the reader, and represents an example of a general strategy for making the compounds described herein.
  • Other methods for preparing the compounds described herein will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples.
  • Compounds of structure 4 can be prepared from precursors of structure 1 by protection of the diol, reduction of the carboxyl to the corresponding aldehyde (structure 3), acetalation, and then deprotection.
  • Compounds of structure 5 can be obtained by acylation or further acetalation of the compounds of structure 4.
  • Compounds of structure 6 can be prepared from precursors of structure 1 by esterification followed by acetalation.
  • Compounds of structure 7 can be prepared by direct acetalation of precursors of structure 1.
  • Compounds of structure 8 can be obtained from precursors of structure 1 by lactonization followed by acetalation.
  • Compounds of structure 9 can be obtained by direct reduction of precursors of structure 1.
  • Compounds of structures 10 and 11 can be prepared from compounds of structure 9 by treatment with formaldehyde.
  • a daily dose may be from about 0.25 mg/kg to about 120 mg/kg or more of body weight, from about 0.5 mg/kg or less to about 70 mg/kg, from about 1.0 mg/kg to about 50 mg/kg of body weight, or from about 1.5 mg/kg to about 10 mg/kg of body weight
  • the dosage range would be from about 17 mg per day to about 8000 mg per day, from about 35 mg per day or less to about 7000 mg per day or more, from about 70 mg per day to about 6000 mg per day, from about 100 mg per day to about 5000 mg per day, or from about 200 mg to about 3000 mg per day.
  • the amount of active compound administered will, of course, be dependent on the subject and disease state being treated, the severity of the affliction, the manner and schedule of administration and the judgment of the prescribing physician.
  • Administration of the compounds disclosed herein or the pharmaceutically acceptable salts thereof can be via any of the accepted modes of administration for agents that serve similar utilities including, but not limited to, orally, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarilly, vaginally, rectally, or intraocularly. Oral and parenteral administrations are customary in treating the indications that are the subject of the preferred embodiments.
  • compositions comprising: (a) a safe and therapeutically effective amount of a compound described herein (including enantiomers, diastereoisomers, tautomers, polymorphs, and solvates thereof), or pharmaceutically acceptable salts thereof; and (b) a pharmaceutically acceptable carrier, diluent, excipient or combination thereof.
  • compositions containing a pharmaceutically-acceptable carrier include compositions containing a pharmaceutically-acceptable carrier.
  • pharmaceutically-acceptable carrier or“pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. In addition, various adjuvants such as are commonly used in the art may be included. Considerations for the inclusion of various components in pharmaceutical compositions are described, e.g., in Gilman et al. (Eds.) (1990); Goodman and Gilman’s: The Pharmacological Basis of Therapeutics, 8th Ed., Pergamon Press, which is incorporated herein by reference in its entirety.
  • substances which can serve as pharmaceutically- acceptable carriers or components thereof, are sugars, such as lactose, glucose and sucrose; starches, such as com starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and methyl cellulose; powdered tragacanth; malt; gelatin; talc; solid lubricants, such as stearic acid and magnesium stearate; calcium sulfate; vegetable oils, such as peanut oil, cottonseed oil, sesame oil, olive oil, com oil and oil of theobroma; polyols such as propylene glycol, glycerine, sorbitol, mannitol, and polyethylene glycol; alginic acid; emulsifiers, such as the TWEENS; wetting agents, such sodium lauryl sulfate; coloring agents; flavoring agents; tableting agents, stabilizers; antioxidants; preservatives;
  • a pharmaceutically-acceptable carrier to be used in conjunction with the subject compound is basically determined by the way the compound is to be administered.
  • compositions described herein are preferably provided in unit dosage form.
  • a "unit dosage form" is a composition containing an amount of a compound that is suitable for administration to an animal, preferably mammal subject, in a single dose, according to good medical practice.
  • the preparation of a single or unit dosage form does not imply that the dosage form is administered once per day or once per course of therapy.
  • Such dosage forms are contemplated to be administered once, twice, thrice or more per day and may be administered as infusion over a period of time (e.g., from about 30 minutes to about 2-6 hours), or administered as a continuous infusion, and may be given more than once during a course of therapy, though a single administration is not specifically excluded.
  • the skilled artisan will recognize that the formulation does not specifically contemplate the entire course of therapy and such decisions are left for those skilled in the art of treatment rather than formulation.
  • compositions described above may be in any of a variety of suitable forms for a variety of routes for administration, for example, for oral, nasal, rectal, topical (including transdermal), ocular, intracerebral, intracranial, intrathecal, intra-arterial, intravenous, intramuscular, or other parental routes of administration.
  • routes for administration for example, for oral, nasal, rectal, topical (including transdermal), ocular, intracerebral, intracranial, intrathecal, intra-arterial, intravenous, intramuscular, or other parental routes of administration.
  • oral and nasal compositions comprise compositions that are administered by inhalation, and made using available methodologies.
  • a variety of pharmaceutically-acceptable carriers well-known in the art may be used.
  • Pharmaceutically-acceptable carriers include, for example, solid or liquid fillers, diluents, hydrotropies, surface-active agents, and encapsulating substances.
  • Optional pharmaceutically-active materials may be included, which do not substantially interfere with the inhibitory activity of the compound.
  • the amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound.
  • Various oral dosage forms can be used, including such solid forms as tablets, capsules, granules and bulk powders. Tablets can be compressed, tablet triturates, enteric-coated, sugar-coated, film-coated, or multiple-compressed, containing suitable binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, flow- inducing agents, and melting agents.
  • Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules, and effervescent preparations reconstituted from effervescent granules, containing suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, melting agents, coloring agents and flavoring agents.
  • the pharmaceutically-acceptable carrier suitable for the preparation of unit dosage forms for peroral administration is well-known in the art.
  • Tablets typically comprise conventional pharmaceutically-compatible adjuvants as inert diluents, such as calcium carbonate, sodium carbonate, mannitol, lactose and cellulose; binders such as starch, gelatin and sucrose; disintegrants such as starch, alginic acid and croscarmelose; lubricants such as magnesium stearate, stearic acid and talc.
  • Glidants such as silicon dioxide can be used to improve flow characteristics of the powder mixture.
  • Coloring agents such as the FD&C dyes, can be added for appearance.
  • Sweeteners and flavoring agents such as aspartame, saccharin, menthol, peppermint, and fruit flavors, are useful adjuvants for chewable tablets.
  • Capsules typically comprise one or more solid diluents disclosed above.
  • the selection of carrier components depends on secondary considerations like taste, cost, and shelf stability, which are not critical, and can be readily made by a person skilled in the art.
  • Peroral compositions also include liquid solutions, emulsions, suspensions, and the like.
  • the pharmaceutically-acceptable carriers suitable for preparation of such compositions are well known in the art.
  • Typical components of carriers for syrups, elixirs, emulsions and suspensions include ethanol, glycerol, propylene glycol, polyethylene glycol, liquid sucrose, sorbitol and water.
  • typical suspending agents include methyl cellulose, sodium carboxymethyl cellulose, AVICEL RC-591, tragacanth and sodium alginate;
  • typical wetting agents include lecithin and polysorbate 80;
  • typical preservatives include methyl paraben and sodium benzoate.
  • Peroral liquid compositions may also contain one or more components such as sweeteners, flavoring agents and colorants disclosed above.
  • compositions may also be coated by conventional methods, typically with pH or time-dependent coatings, such that the subject compound is released in the gastrointestinal tract in the vicinity of the desired topical application, or at various times to extend the desired action.
  • dosage forms typically include, but are not limited to, one or more of cellulose acetate phthalate, polyvinylacetate phthalate, hydroxypropyl methyl cellulose phthalate, ethyl cellulose, Eudragit coatings, waxes and shellac.
  • compositions described herein may optionally include other drug actives.
  • compositions useful for attaining systemic delivery of the subject compounds include sublingual, buccal and nasal dosage forms.
  • Such compositions typically comprise one or more of soluble filler substances such as sucrose, sorbitol and mannitol; and binders such as acacia, microcrystalline cellulose, carboxymethyl cellulose and hydroxypropyl methyl cellulose. Glidants, lubricants, sweeteners, colorants, antioxidants and flavoring agents disclosed above may also be included.
  • a liquid composition which is formulated for topical ophthalmic use, is formulated such that it can be administered topically to the eye.
  • the comfort should be maximized as much as possible, although sometimes formulation considerations (e.g. drug stability) may necessitate less than optimal comfort.
  • the liquid should be formulated such that the liquid is tolerable to the patient for topical ophthalmic use.
  • an ophthalmically acceptable liquid should either be packaged for single use, or contain a preservative to prevent contamination over multiple uses.
  • solutions or medicaments are often prepared using a physiological saline solution as a major vehicle. Ophthalmic solutions should preferably be maintained at a comfortable pH with an appropriate buffer system.
  • the formulations may also contain conventional, pharmaceutically acceptable preservatives, stabilizers and surfactants.
  • Preservatives that may be used in the pharmaceutical compositions disclosed herein include, but are not limited to, benzalkonium chloride, PHMB, chlorobutanol, thimerosal, phenylmercuric, acetate and phenylmercuric nitrate.
  • a useful surfactant is, for example, Tween 80.
  • various useful vehicles may be used in the ophthalmic preparations disclosed herein. These vehicles include, but are not limited to, polyvinyl alcohol, povidone, hydroxypropyl methyl cellulose, poloxamers, carboxymethyl cellulose, hydroxyethyl cellulose and purified water.
  • Tonicity adjustors may be added as needed or convenient They include, but are not limited to, salts, particularly sodium chloride, potassium chloride, mannitol and glycerin, or any other suitable ophthalmically acceptable tonicity adjustor.
  • buffers include acetate buffers, citrate buffers, phosphate buffers and borate buffers. Acids or bases may be used to adjust the pH of these formulations as needed.
  • an ophthalmically acceptable antioxidant includes, but is not limited to, sodium metabisulfite, sodium thiosulfate, acetylcysteine, butylated hydroxyanisole and butylated hydroxytoluene.
  • excipient components which may be included in the ophthalmic preparations, are chelating agents.
  • a useful chelating agent is edetate disodium, although other chelating agents may also be used in place or in conjunction with it
  • Topical formulations may generally be comprised of a pharmaceutical carrier, co-solvent, emulsifier, penetration enhancer, preservative system, and emollient
  • a pharmaceutically acceptable diluent such as a saline or dextrose solution.
  • Suitable excipients may be included to achieve the desired pH, including but not limited to NaOH, sodium carbonate, sodium acetate, HC1, and citric acid.
  • the pH of the final composition ranges from 2 to 8, or preferably from 4 to 7.
  • Antioxidant excipients may include sodium bisulfite, acetone sodium bisulfite, sodium formaldehyde, sulfoxylate, thiourea, and EDTA.
  • Other non-limiting examples of suitable excipients found in the final intravenous composition may include sodium or potassium phosphates, citric acid, tartaric acid, gelatin, and carbohydrates such as dextrose, mannitol, and dextran.
  • Antimicrobial agents may also be included to achieve a bacteriostatic or fungistatic solution, including but not limited to phenylmercuric nitrate, thimerosal, benzethonium chloride, benzalkonium chloride, phenol, cresol, and chlorobutanol.
  • compositions for intravenous administration may be provided to caregivers in the form of one more solids that are reconstituted with a suitable diluent such as sterile water, saline or dextrose in water shortly prior to administration
  • a suitable diluent such as sterile water, saline or dextrose in water shortly prior to administration
  • the compositions are provided in solution ready to administer parenterally.
  • the compositions are provided in a solution that is further diluted prior to administration.
  • the combination may be provided to caregivers as a mixture, or the caregivers may mix the two agents prior to administration, or the two agents may be administered separately.
  • Some embodiments of the present invention include methods of treating a liver disease, disorder, or condition by administering the compounds or pharmaceutical compositions described herein to a subject in need thereof.
  • the liver disease, disorder, or condition involves the metabolism, storage, or homeostatic control of biochemical end products such as cholesterol, fatty acids, bile, triglycerides, plasma proteins, carrier proteins, lipoproteins, acute phase proteins, apolipoproteins, carbohydrates, hemostatic factors, fibrinolysis factors, thrombopoietin, insulin-like growth factor 1, hepcidin, angiotensinogen, vitamin A, vitamin D, vitamin B12, vitamin K, iron, copper, or catalase.
  • biochemical end products such as cholesterol, fatty acids, bile, triglycerides, plasma proteins, carrier proteins, lipoproteins, acute phase proteins, apolipoproteins, carbohydrates, hemostatic factors, fibrinolysis factors, thrombopoietin, insulin-like growth factor 1, hepcidin, an
  • the liver disease, disorder, or condition being treated is liver fibrosis, cirrhosis, hepatitis, alcoholic liver disease, fatty liver, liver cancer, hemochromatosis, Wilson’s disease, primary sclerosing cholangitis, alpha 1-antitrypsin deficiency, hyperglycemia, hypoglycemia, or diabetes.
  • Some embodiments of the present invention include methods of treating a cardiovascular disease, disorder, or condition, by administering the compounds or pharmaceutical compositions described herein to a subject in need thereof.
  • the cardiovascular disease, disorder, or condition is dyslipidemia or atherosclerosis.
  • the cardiovascular disease, disorder, or condition is hyperlipidemia.
  • Some embodiments of the present invention include methods of altering blood lipid concentration by administering the compounds or pharmaceutical compositions described herein to a subject in need thereof.
  • Some embodiments of the present invention include methods of inhibiting HMG-CoA reductase by administering the compounds or pharmaceutical compositions described herein to a subject in need thereof.
  • Some embodiments of the present invention include methods of reducing the side effects of liver-targeting or liver-activated drugs by administering the compounds or pharmaceutical compositions described herein, rather than their corresponding active drag compound, to a subject in need thereof.
  • the corresponding active drug compound is a statin.
  • side effects that can be reduced include headache, difficulty sleeping, flushing of the skin, muscle aches, muscle tenderness, muscle weakness, drowsiness, dizziness, nausea, vomiting, abdominal pain, bloating, gas, diarrhea, constipation, or rash.
  • Some embodiments of the present invention include methods of increasing the therapeutic index of liver-targeting or liver-activated therapeutic agents.
  • the subject is a mammal.
  • the subject is a human.
  • Further embodiments include administering a combination of compounds to a subject in need thereof.
  • a combination can include a compound or pharmaceutical composition described herein with an additional medicament.
  • Some embodiments include co-administering a compound or pharmaceutical composition described herein, with an additional medicament.
  • co-administration it is meant that the two or more agents may be found in the patient’s bloodstream at the same time, regardless of when or how they are actually administered.
  • the agents are administered simultaneously.
  • administration in combination is accomplished by combining the agents in a single dosage form.
  • the agents are administered sequentially.
  • the agents are administered through the same route, such as orally.
  • the agents are administered through different routes, such as one being administered orally and another being administered intravenously.
  • the additional medicament is a cholesterol absorption inhibitor, a hypertension medicament, niacin, or aspirin.
  • the cholesterol absorption inhibitor is ezetimibe.
  • the hypertension medicament is a diuretic, a beta-blocker, an angiotensin-converting enzyme inhibitor, an angiotensin receptor blocker, a calcium channel blocker, an alpha blocker, an alpha-2 receptor agonist, a combined alpha and beta-blocker, a central agonist, a peripheral adrenergic inhibitor, or a vasodilator.
  • Example 7 operated according to Scheme I from rosuvastatin and ( ⁇ )-2,4-dihydroxypentane in a manner similar to that of Compound 101. [M+H] + calculated for C27H38FN3O6S: 552.26; found: 552.2.
  • Compoimds 116 was prepared according to Scheme I from rosuvastatin and acetaldehyde. [M+H] + calculated for C24H30FN3O6S: 508.19; found: 508.1.
  • Compounds 125 can be prepared according to a standard procedure from rosuvastatin and chloromethyl ethyl ether. [M+H] + calculated for CasHioFNgOeS: 598.26.
  • Compounds 127 can be prepared according to a standard procedure from rosuvastatin and chloromethyl ethyl ether. [M+H] + calculated for CisHgeFNsOeS: 526.24.
  • the liver specificity of the disclosed compounds were determined and compared with their corresponding active drug compounds.
  • the disclosed compounds and reference active drug compounds were administered at 5-20 mg/kg to fasted rats by oral gavage.
  • Plasma concentrations of the active, metabolite, and the prodrug compounds in circulation and in the hepatic portal vein were determined by a standard HPLC-UV method. Concentrations of the same in the liver, small intestine, and other organs were measured by the standard LC-MS method.
  • Table 1 summarizes the results, which demonstrates improved liver distribution of the disclosed compounds relative to their active drug counterparts.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Diabetes (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Disclosed herein are acetal and cyclic acetal compounds, compositions, their preparation, and their uses. Some embodiments relate to their use as liver-targeting compounds.

Description

ACETAL COMPOUNDS AND THERAPEUTIC USES THEREOF
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of U.S. Provisional Application No. 62/615,357 filed January 9, 2018 entitled“ACETAL COMPOUNDS AND THERAPEUTIC USES THEREOF’, which is incorporated by reference in its entirety.
BACKGROUND
Field
[0002] The present disclosure relates to the field of chemistry and medicine. More specifically, the present disclosure relates to acetal and cyclic acetal compounds, compositions, their preparation, and their use as therapeutic agents.
Description of the Related Art
[0003] Statins are a class of liver-targeting compounds that inhibit HMG-CoA reductase, one of the liver enzymes in the cholesterol biosynthetic pathway. Statins are used as lipid-lowing medicines for the prophylactic and therapeutic treatment of coronary heart disease. Despite their ubiquitous use, statins, like other liver-targeting drugs, suffer from poor oral bioavailability and inadequate distribution to the liver. In fact, a significant portion of patients cannot take statins because of side effects from statin biological activity outside of the liver.
[0004] Thus, there is a need for improved liver-targeting compounds and strategies to increase the bioavailability of liver-targeting drugs, to increase drug distribution to the liver, and to reduce drug biological activity outside of the liver.
SUMMARY
[0005] Some embodiments described herein relate to compounds having the structure of Formula I:
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, and R4 have any of the values described herein.
[0006] In some embodiments, R1 is selected from the group consisting of optionally substituted Ci-galkyl, -COOR5, optionally substituted C2-ioalkoxyalkyl, and optionally substituted 3-12 membered heterocyclyl. Alternatively, in some embodiments, R1 and R2 together with the atoms to which they are attached form an optionally substituted 4-10 membered heterocyclyl, or R1 and R3 together with the atoms to which they are attached form an optionally substituted 6-10 membered heterocyclyl.
[0007] R2 in some embodiments is selected from the group consisting of H, - C(0)R6, and optionally substituted C2-ioalkoxyalkyl. Alternatively, in some embodiments, R1 and R2 together with the atoms to which they are attached form an optionally substituted 4-10 membered heterocyclyl, or R2 and R3 together with the atoms to which they are attached form a 6-10 membered heterocyclyl, optionally substituted with one or more R7.
[0008] In some embodiments, R3 is selected from the group consisting of H, - C(0)R8, and optionally substituted Ca-ioalkoxyalkyl. Alternatively, in some embodiments, R1 and R3 together with the atoms to which they are attached form an optionally substituted 6-10 membered heterocyclyl, or R2 and R3 together with the atoms to which they are attached form a 6-10 membered heterocyclyl, optionally substituted with one or more R7.
[0009] R4 in some embodiments is a fragment of a therapeutic agent.
[0010] In some embodiments, each of R5, R6, and R8 is independently selected from the group consisting of H, optionally substituted Ci-ealkyl, halogen, optionally substituted Ca^alkenyl, optionally substituted C2-6alkenyl, optionally substituted C3- 7carbocyclyl, optionally substituted C6-ioaryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl.
[0011] In some embodiments, each R7 is independently selected from the group consisting of optionally substituted Ci-ealkyl, oxo, optionally substituted C6-isaryl, and optionally substituted 5-18 membered heteroaryl. [0012] In some embodiments, at least one of R2 and R3 is not H, or R1 is not -
COOH. Moreover, in some embodiments, if R1 and R3 together with the atoms to which they are attached form a 6 membered heterocyclyl, then R2 is not H.
[0013] Some embodiments described herein relate to compounds having the structure of Formula P:
Figure imgf000004_0001
[0014] or a pharmaceutically acceptable salt thereof, wherein R37, R38, R39, and Z have any of the values described herein.
[0015] In some embodiments, Z is O, S, or NR40.
[0016] R37 in some embodiments is selected from the group consisting of H, a fragment of a therapeutic agent, optionally substituted C1-8alkyl, Ci-6haloalkyl, Ci- eheteroalkyl, optionally substituted Ca-vcarbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C6-isaiyl, 5-10 membered heteroaryl, and optionally substituted Ca-ioalkoxy alkyl.
[001h In some embodiments, each of R38 and R39 is independently selected from the group consisting of a fragment of a therapeutic agent, optionally substituted Ci-ealkyl, Ci- ehaloalkyl, C1-6heteroalkyl, optionally substituted C3-7carbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C1-6saryl, optionally substituted C2- loalkoxyalkyl, and optionally substituted 5-10 membered heteroaryl.
[0018] In some embodiments, at least one of R37, R38, and R39 is a fragment of a therapeutic agent.
[0019] In some embodiments, the fragment of a therapeutic agent is further optionally substituted with one or more moieties selected from the group consisting of
Figure imgf000005_0001
i-ealkyl, halo, and C6-isaryl. In some such embodiments, each A is independently selected from O, S, and NR43, provided that at least one A is O. In some such embodiments, each of R41, R42, and R43 is independently selected from the group consisting of H, optionally substituted Ci^alkyl, Ci-ehaloalkyl, Ci- eheteroalkyl, optionally substituted C3-7carbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C6-isaryl, optionally substituted Cz-ioalkoxy alkyl, and optionally substituted 5-10 membered heteroaryl.
[0020] In some embodiments, R40 is independently selected from the group consisting of optionally substituted Ci-ealkyl, Ci-ehaloalkyl, Ci-eheteroalkyl, optionally substituted C3-7carbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C6-isaryl, optionally substituted C2-ioalkoxyalkyl, and optionally substituted 5-10 membered heteroaryl.
DETAILED DESCRIFTION
[0021] Compoimds of Formula I and II
[0022] In some embodiments, liver-targeting compounds containing an acetal moiety are provided that afford increased bioavailability relative to their active drag counterparts, increased drug distribution to the liver relative to their active drug counterparts, and/or reduced biological activity outside of the liver.
[0023] Various embodiments of these compounds include compounds having the structures of Formula I or P as described above or pharmaceutically acceptable salts thereof. In some embodiments, the compounds of Formula I are also represented by the structure of Formula la, lb, Ic, Id, Ie, If, or Ig:
Figure imgf000006_0001
Ig , or pharmaceutically acceptable salts thereof, wherein Rl, R2, R3, R4, R5, R6, R7, R8, R9, RIO, Rl 1, R12, R13, R14, R15, R16, R17, R18, R19, R20, R21, R22, R23, R24, R25, R26, R27, R28, R29, R30, R31, R32, R33, R34, R35, R36, X, and Y have any of the values described herein.
[0024] In some embodiments of the compounds of Formula Ic, X is an optionally substituted Cl-7alkylene linker. In some such embodiments, X is a Cl-7alkylene linker optionally substituted with one or more Rl l. In some such embodiments, Rl l is selected from the group consisting of Cl-6alkyl, Cl-6alkenyl, Cl-6alkynyl, Cl-6heteroalkyl, C3- 7carbocyclyl, 3-10 member ed heterocyclyl, C6-18aryl, C6-18arylCl-6alkyl, 5-10 membered heteroaryl, 5-10 membered heteroarylCl-6alkyl, halo, cyano, hydroxy, Cl-6alkoxy, C2- lOalkoxyalkyl, C6-18aiyloxy, sulfhydryl, haloCl-6alkyl, haloCl-6alkoxy, Cl-6alkylthio, C6-18arylthio, and nitro. In some such embodiments, Rll is selected from the group consisting of Cl-6alkyl, C3-7carbocyclyl, Cl-6alkoxy, C6-18aryl, or halo. In some further embodiments, X is a C2alkylene linker. In some particular embodiments, X is -CH2CH2-. In some further embodiments, X is a C3alkylene linker. In some particular embodiments, X is - CH2CH2CH2-. In some particular embodiments, X is -CH2(CH2CH3)CH2-. In other particular embodiments, X is -CH2(CH3)CH2CH2(CH3)-.
[0025] In some embodiments of the compounds of Formula Id, each of R9 and
RIO is independently an optionally substituted Cl-6alkyl. In some such embodiments, each of R9 and RIO is independently a Cl-6alkyl optionally substituted with one or more R12. In some such embodiments, R12 is selected from the group consisting of Cl-6alkyl, Cl- 6alkenyl, Cl-6alkynyl, Cl-6heteroalkyl, C3-7carbocyclyl, C3-7carbocyclylCl-6alkyl, 3-10 membered heterocyclyl, 3-10 membered heterocyclylCl-6alkyl, C6-18aryl, C6-18arylCl- 6alkyl, 5-10 membered heteroaryl, 5-10 membered heteroarylCl-6alkyl, and haloCl-6alkyl. In some further embodiments, R9 is a Cl-6alkyl. In some particular embodiments, R9 is ethyl. In some further embodiments, R10 is a Cl-6alkyl. In some particular embodiments, R10 is ethyl.
[0026] In some embodiments of the compounds Formula If, Y is a Cl-5alkylene linker, optionally substituted with one or more R7. In some such embodiments, R7 is selected from the group consisting of optionally substituted Cl-8alkyl, oxo, optionally substituted C6-18aryl, and optionally substituted 5-18 membered heteroaryl. In some further embodiments, Y is a Clalkylene linker.
[002h In some embodiments of the compounds of Formula I, la, lb, or If, R1 is an optionally substituted Cl-8alkyl. In some such embodiments, R1 is a Cl-8alkyl optionally substituted with one or more R13. In some such embodiments, R13 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro. In some particular embodiments, R1 is -CH20H.
[0028] In some embodiments of the compounds of Formula I, la, lb, or If, R1 is - COOR5. In some such embodiments, R5 is H. In other such embodiments, R5 is methyl. [0029] In some embodiments of the compounds of Formula I, la, lb, or If, R1 is an optionally substituted C2-10alkoxyalkyl. In some such embodiments, R1 is a C2- lOalkoxyalkyl optionally substituted with one or more R14. In some such embodiments, R14 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro. In some particular embodiments, R1 is -CH20CH20CH2CH3. In some particular embodiments, R1 is - CH(OCH2CH3)2.
[0030] In some embodiments of the compounds of Formula I, la, lb, or If, R1 is an optionally substituted 3-12 membered heterocyclyl. In some such embodiments, R1 is a 3-12 membered heterocyclyl optionally substituted with one or more R15. In some such embodiments, R15 is selected from the group consisting of Cl-6alkyl, Cl-6alkenyl, Cl- 6alkynyl, Cl-6heteroalkyl, C3-7carbocyclyl, 3-10 membered heterocyclyl, C6-18aryl, C6- 18arylCl-6alkyl, 5-10 membered heteroaryl, 5-10 membered heteroarylCl-6alkyl, halo, cyano, hydroxy, Cl-6alkoxy, C2-10alkoxyalkyl, aryloxy, sulfhydryl, haloCl-6alkyl, haloCl- 6alkoxy, Cl-6alkylthio, C6-18arylthio, and nitro. In some further embodiments, R1 is a 5
membered heterocyclyl. In some particular embodiments, R1 is
Figure imgf000008_0001
or
Figure imgf000008_0002
In some further embodiments, R1 is a 6 membered heterocyclyl. In some
particular embodiments, R1 is
Figure imgf000008_0003
[0031] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, or Ie,
R2 is H.
[0032] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, Ie, or
Ig, R2 is -C(0)R6. In some such embodiments, R6 is an optionally substituted Cl-6alkyl. In some such embodiments, R6 is a Cl-6alkyl optionally substituted with one or more R16. In some such embodiments, R16 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6- lOarylthio, and nitro. In some particular embodiments, R6 is -CH(CH3)2.
[0033] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, Ie, or Ig, R2 is an optionally substituted C2-10alkoxyalkyl. In some such embodiments, R2 is a C2-10alkoxyalkyl optionally substituted with one or more R17. In some such embodiments, R17 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6-10aryloxy, sulfhydiyl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro. In some particular embodiments, R2 is -CH20CH2CH3.
[0034] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, or Ie,
R3 is H.
[0035] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, or Ie, R3 is -C(0)R8. In some such embodiments, R8 is an optionally substituted Cl-6alkyl. In some such embodiments, R8 is a Cl-6alkyl optionally substituted with one or more R18. In some such embodiments, R18 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6- lOarylthio, and nitro. In some particular embodiments, R8 is -CH(CH3)2.
[0036] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, or Ie, R3 is an optionally substituted C2-10alkoxyalkyl. In some such embodiments, R3 is a C2- lOalkoxy alkyl optionally substituted with one or more R19. In some such embodiments, R19 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6- lOaryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro. In some particular embodiments, R3 is -CH20CH2CH3.
[003h In some embodiments of the compounds of Formula I, la, lb, Ic, Id, Ie, or
If, R7 is an optionally substituted Cl-8alkyl. In some such embodiments, R7 is a Cl-8alkyl optionally substituted with one or more R20. In some such embodiments, R20 is selected from the group consisting of halo, cyano, hydroxy, Cl-6alkoxy, C6-10aryl, C6-10aryloxy, sulfhydryl, haloCl-6alkoxy, Cl-6alkylthio, C6-10arylthio, and nitro. In some particular embodiments, R7 is methyl. In some other particular embodiments, R7 is -CH2CH2CH20H.
[0038] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, Ie, or If, R7 is oxo. [0039] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, Ie, or If, R7 is an optionally substituted C6-18aryl. In some such embodiments, R7 is a C6-18aryl optionally substituted with one or more R21. In some such embodiments, R21 is selected from the group consisting of Cl-6alkyl, halo, cyano, hydroxy, Cl-6alkoxy, haloCl-6alkyl, haloCl-6alkoxy, nitro, -C(=0)NR22R23, -N(R24)C(=0)R25, -C(=0)0R26, -0C(=0)R27, and -C(=0)R28. In some such embodiments, each of R22, R23, R24, R25, R26, R27, and R28 is independently selected from the group consisting of hydrogen, Cl-6alkyl, C2- 6alkenyl, C2-6alkynyl, C3-7carbocyclyl, C6-10aiyl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl. In some further embodiments, R7 is a phenyl. In some particular
embodiments, R7 is
Figure imgf000010_0001
[0040] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, Ie, or If, R7 is an optionally substituted 5-18 membered heteroaryl. In some such embodiments, R7 is a 5-18 membered heteroaryl optionally substituted with one or more R29. In some such embodiments, R29 is selected from the group consisting of Cl-6alkyl, halo, cyano, hydroxy, Cl-6alkoxy, haloCl-6alkyl, haloCl-6alkoxy, nitro, -C(=O)NR30R31, -
N(R32)C(=0)R33, -C(=0)0R34, -0C(=0)R35, and -C(=0)R36. In some such embodiments, each of R30, R31, R32, R33, R34, R35, and R36 is independently selected from the group consisting of hydrogen, Cl-6alkyl, C2-6alkenyl, C2-6alkynyl, C3- 7carbocyclyl, C6-10aryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl. In some further embodiments, R7 is a 6 membered heteroaryl. In some particular embodiments,
Figure imgf000010_0002
[0041] In some embodiments of the compounds of Formula I, la, lb, Ic, Id, Ie, If, or Ig, R4 is a fragment of a liver-targeting therapeutic agent or a fragment of a liver-activated therapeutic agent. In some such embodiments, R4 is a fragment of a lipid-lowering therapeutic agent or a fragment of a cholesterol-biosynthesis-inhibiting therapeutic agent In some such embodiments, R4 is a fragment of a HMG-CoA reductase inhibitor. In some such embodiments, R4 is a fragment of a statin. In some further embodiments, R4 is selected from the group consisting of
Figure imgf000011_0002
Figure imgf000011_0001
In some particular embodiments, R4 is
Figure imgf000012_0001
[0042] In some embodiments of the compounds of Formula P, Z is O.
[0043] In some embodiments, R37 is H.
[0044] R39 in some embodiments is ethyl.
[0045] R38 in some embodiments is the optionally substituted fragment of a therapeutic agent. In some such embodiments, the fragment of a therapeutic agent is
optionally substituted with one or
Figure imgf000012_0002
more moieties Cl-6alkyl, halo, and C6-18aryl. In some such embodiments, the substituent is m R38 has the structure
In some embodiments, the
Figure imgf000012_0003
fragment of the therapeutic agent is
Figure imgf000013_0001
optionally substituted with
Figure imgf000013_0002
halo, and C6-18aryl. In some such embodiments, the substituent is
Figure imgf000013_0004
H
, such that R38 has the structure
Figure imgf000013_0003
[0046] In some embodiments, A is O.
[0047] In some embodiments, R41 is H.
[0048] In some embodiments, R42 is ethyl.
[0049] In some embodiments, the compounds of Formula I, la, lb, Ic, Id, Ie, If, Ig, or P as described herein are selected from the group consisting of
Figure imgf000013_0005
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
[0050] In some embodiments, the pharmaceutically acceptable salts are selected from alkaline metal salts or ammonium salts. In some embodiments, the pharmaceutically acceptable salts are sodium salts, including disodium salts.
[0051] Where the compounds disclosed herein have at least one chiral center, they may exist as individual enantiomers and diastereomers or as mixtures of such isomers, including racemates. Separation of the individual isomers or selective synthesis of the individual isomers is accomplished by application of various methods which are well known to practitioners in the art Unless otherwise indicated, all such isomers and mixtures thereof are included in the scope of the compounds disclosed herein. Furthermore, compounds disclosed herein may exist in one or more crystalline or amorphous forms. Unless otherwise indicated, all such forms are included in the scope of the compounds disclosed herein, including any polymorphic forms. In addition, some of the compounds disclosed herein may form solvates with water (i.e., hydrates) or common organic solvents. Unless otherwise indicated, such solvates are included in the scope of the compounds disclosed herein.
[0052] The skilled artisan will recognize that some structures described herein may be resonance forms or tautomers of compounds that may be fairly represented by other chemical structures, even when kinetically; the artisan recognizes that such structures may only represent a very small portion of a sample of such compound(s). Such compounds are considered within the scope of the structures depicted, though such resonance forms or tautomers are not represented herein.
[0053] Isotopes may be present in the compounds described. Each chemical element as represented in a compound structure may include any isotope of said element. For example, in a compound structure a hydrogen atom may be explicitly disclosed or understood to be present in the compound. At any position of the compound that a hydrogen atom may be present, the hydrogen atom can be any isotope of hydrogen, including but not limited to hydrogen- 1 (protium) and hydrogen-2 (deuterium). Thus, reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise.
Definitions
[0054] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. All patents, applications, published applications, and other publications referenced herein are incorporated by reference in their entirety unless stated otherwise. In the event that there are a plurality of definitions for a term herein, those in this section prevail unless stated otherwise. As used in the specification and the appended claims, the singular forms“a,” “an,” and“the” include plural referents unless the context clearly dictates otherwise. Unless otherwise indicated, conventional methods of mass spectroscopy, NMR, HPLC, protein chemistry, biochemistry, recombinant DNA techniques, and pharmacology are employed. The use of“or” or“and" means“and/or" unless stated otherwise. Furthermore, use of the term“including” as well as other forms, such as“include",“includes,” and“included,” is not limiting. As used in this specification, whether in a transitional phrase or in the body of the claim, the terms“comprise(s)” and“comprising” are to be interpreted as having an open- ended meaning. That is, the terms are to be interpreted synonymously with the phrases “having at least” or“including at least." When used in the context of a process, the term “comprising” means that the process includes at least the recited steps, but may include additional steps. When used in the context of a compound, composition, or device, the term “comprising” means that the compound, composition, or device includes at least the recited features or components, but may also include additional features or components.
[0055] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
[0056] As used herein, common organic abbreviations are defined as follows:
Figure imgf000018_0001
Figure imgf000019_0001
[005h As used herein,“Ca to Cb” or“Ca-b” in which“a” and“b” are integers refer to the number of carbon atoms in the specified group. That is, the group can contain from “a” to“b”, inclusive, carbon atoms. Thus, for example, a“Ci to Ct alkyl” or“CM alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH3-, CH3CH2-, CH3CH2CH2-, (CH3)2CH-, CH3CH2CH2CH2-, CH3CH2CH(CH3)- and (C¾)3C-.
[0058] The term“halogen” or“halo,” as used herein, means any one of the radiostable atoms of column 7 of the Periodic Table of the Elements, e.g., fluorine, chlorine, bromine, or iodine, with fluorine and chlorine being preferred.
[0059] As used herein,“alkyl” refers to a straight or branched hydrocarbon chain that is fully saturated (i.e., contains no double or triple bonds). The alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as“1 to 20” refers to each integer in the given range; e.g.,“1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term“alkyl” where no numerical range is designated). The alkyl group may also be a medium size alkyl having 1 to 9 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 4 carbon atoms. The alkyl group may be designated as“CM alkyl” or similar designations. By way of example only,“CM alkyl” indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from the group consisting of methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, and the like.
[0060] As used herein,“alkoxy” refers to the formula -OR wherein R is an alkyl as is defined above, such as“C1-9 alkoxy”, including but not limited to methoxy, ethoxy, n- propoxy, l-methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, and tert-butoxy, and the like.
[0061] As used herein,“alkylthio” refers to the formula -SR wherein R is an alkyl as is defined above, such as“C1-9 alkylthio” and the like, including but not limited to methylmercapto, ethylmercapto, n-propylmercapto, 1 -methylethy lmercapto
(isopropylmercapto), n-butylmercapto, iso-butylmercapto, sec-butylmercapto, tert- butylmercapto, and the like.
[0062] As used herein,“alkenyl” refers to a straight or branched hydrocarbon chain containing one or more double bonds. The alkenyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term“alkenyl” where no numerical range is designated. The alkenyl group may also be a medium size alkenyl having 2 to 9 carbon atoms. The alkenyl group could also be a lower alkenyl having 2 to 4 carbon atoms. The alkenyl group may be designated as “C2-* alkenyl” or similar designations. By way of example only,“C2-4 alkenyl” indicates that there are two to four carbon atoms in the alkenyl chain, i.e., the alkenyl chain is selected from the group consisting of ethenyl, propen-l-yl, propen-2-yl, propen-3-yl, buten-l-yl, buten-2-yl, buten-3-yl, buten- 4-yl, 1 -methyl-propen- 1 -yl, 2-methyl-propen- 1 -yl, 1-ethyl-ethen-l-yl, 2-methyl-propen-3-yl, buta-l,3-dienyl, buta-l,2,-dienyl, and buta-l,2-dien-4-yl. Typical alkenyl groups include, but are in no way limited to, ethenyl, propenyl, butenyl, pentenyl, and hexenyl, and the like. [0063] As used herein,“alkynyl” refers to a straight or branched hydrocarbon chain containing one or more triple bonds. The alkynyl group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term“alkynyl” where no numerical range is designated. The alkynyl group may also be a medium size alkynyl having 2 to 9 carbon atoms. The alkynyl group could also be a lower alkynyl having 2 to 4 carbon atoms. The alkynyl group may be designated as “C2-4 alkynyl” or similar designations. By way of example only,“C2-4 alkynyl” indicates that there are two to four carbon atoms in the alkynyl chain, i.e., the alkynyl chain is selected from the group consisting of ethynyl, propyn-l-yl, propyn-2-yl, butyn-l-yl, butyn-3-yl, butyn-4-yl, and 2- butynyl. Typical alkynyl groups include, but are in no way limited to, ethynyl, propynyl, butynyl, pentynyl, and hexynyl, and the like.
[0064] As used herein,“heteroalkyl” refers to a straight or branched hydrocarbon chain containing one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur, in the chain backbone. The heteroalkyl group may have 1 to 20 carbon atom, although the present definition also covers the occurrence of the term“heteroalkyl” where no numerical range is designated. The heteroalkyl group may also be a medium size heteroalkyl having 1 to 9 carbon atoms. The heteroalkyl group could also be a lower heteroalkyl having 1 to 4 carbon atoms. The heteroalkyl group may be designated as“Ci- heteroalkyl” or similar designations. The heteroalkyl group may contain one or more heteroatoms. By way of example only,“CM heteroalkyl” indicates that there are one to four carbon atoms in the heteroalkyl chain and additionally one or more heteroatoms in the backbone of the chain. In various embodiments, a heteroalkyl may contain from 1 to 10 heteroatoms, for example, 1 to 5 heteroatoms, 1 to 3 heteroatoms, 2 heteroatoms, or 1 heteroatom.
[0065] As used herein,“alkylene” means a branched, or straight chain fully saturated di-radical chemical group containing only carbon and hydrogen that is attached to the rest of the molecule via two points of attachment (i.e., an alkanediyl). The alkylene group may have 1 to 20 carbon atoms, although the present definition also covers the occurrence of the term alkylene where no numerical range is designated. The alkylene group may also be a medium size alkylene having 1 to 9 carbon atoms. The alkylene group could also be a lower alkylene having 1 to 4 carbon atoms. The alkylene group may be designated as“Ci-4 alkylene” or similar designations. By way of example only,“C1-4 alkylene” indicates that there are one to four carbon atoms in the alkylene chain, i.e., the alkylene chain is selected from the group consisting of methylene, ethylene, ethan- 1,1-diyl, propylene, propan-
1.1-diyl, propan-2, 2-diyl, 1 -methyl-ethylene, butylene, butan- 1,1-diyl, butan-2,2-diyl, 2- methy 1-propan- 1,1-diyl, 1 -methyl-propylene, 2-methyl-propylene, 1,1 -dimethyl-ethylene,
1.2-dimethyl-ethylene, and 1 -ethyl-ethylene.
[0066] As used herein,“alkenylene” means a straight or branched chain di-radical chemical group containing only carbon and hydrogen and containing at least one carbon- carbon double bond that is attached to the rest of the molecule via two points of attachment. The alkenylene group may have 2 to 20 carbon atoms, although the present definition also covers the occurrence of the term alkenylene where no numerical range is designated. The alkenylene group may also be a medium size alkenylene having 2 to 9 carbon atoms. The alkenylene group could also be a lower alkenylene having 2 to 4 carbon atoms. The alkenylene group may be designated as“C2-4 alkenylene” or similar designations. By way of example only,“C2-4 alkenylene” indicates that there are two to four carbon atoms in the alkenylene chain, i.e., the alkenylene chain is selected from the group consisting of ethenylene, ethen-l,l-diyl, propenylene, propen- 1,1-diyl, prop-2-en-l,l-diyl, 1-methyl- ethenylene, but-l-enylene, but-2-enylene, but-l,3-dienylene, buten- 1,1-diyl, but-l,3-dien- 1,1-diyl, but-2-en- 1,1-diyl, but-3-en- 1,1-diyl, l-methyl-prop-2-en-l,l-diyl, 2-methyl-prop-2- en-l,l-diyl, 1-ethyl-ethenylene, 1,2-dimethyl-ethenylene, 1-methyl-propenylene, 2-methyl- propenylene, 3-methyl-propenylene, 2-methyl-propen- 1,1 -diyl, and 2,2-dimethyl-ethen-l,l- diyl.
[006h The term“aromatic” refers to a ring or ring system having a conjugated pi electron system and includes both carbocyclic aromatic (e.g., phenyl) and heterocyclic aromatic groups (e.g., pyridine). The term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of atoms) groups provided that the entire ring system is aromatic.
[0068] As used herein,“aryl” refers to an aromatic ring or ring system (i.e., two or more fused rings that share two adjacent carbon atoms) containing only carbon in the ring backbone. When the aryl is a ring system, every ring in the system is aromatic. The aryl group may have 6 to 18 carbon atoms, although the present definition also covers the occurrence of the term “aryl” where no numerical range is designated. In some embodiments, the aryl group has 6 to 10 carbon atoms. The aryl group may be designated as “C6-io aryl,”“C6 or Cio aryl,” or similar designations. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, azulenyl, and anthracenyl.
[0069] As used herein,“aiyloxy” and“arylthio” refers to RO- and RS-, in which R is an aryl as is defined above, such as“C6-io aryloxy” or“C6-io arylthio” and the like, including but not limited to phenyloxy.
[0070] An“aralkyl” or“arylalkyl” is an aryl group connected, as a substituent, via an alkylene group, such as“C7-14 aralkyl” and the like, including but not limited to benzyl, 2-phenylethyl, 3-phenylpropyl, and naphthylalkyl. In some cases, the alkylene group is a lower alkylene group (i.e., a CM alkylene group).
[0071] As used herein,“heteroaryl” refers to an aromatic ring or ring system (i.e., two or more fused rings that share two adjacent atoms) that contain(s) one or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur, in the ring backbone. When the heteroaryl is a ring system, every ring in the system is aromatic. The heteroaryl group may have 5-18 ring members (i.e., the number of atoms making up the ring backbone, including carbon atoms and heteroatoms), although the present definition also covers the occurrence of the term“heteroaryl” where no numerical range is designated. In some embodiments, the heteroaryl group has 5 to 10 ring members or 5 to 7 ring members. The heteroaryl group may be designated as “5-7 member ed heteroaryl,”“5-10 membered heteroaryl,” or similar designations. In some embodiments, of the ring members, from 1 to 5 are heteroatoms, for example, the heteroaryl may contain from 1 to 4 heteroatoms, from 1 to 3 heteroatoms, two heteroatoms, or one heteroatom. Examples of heteroaryl rings include, but are not limited to, furyl, thienyl, phthalazinyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, quinolinyl, isoquinlinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, indolyl, isoindolyl, and benzothienyl.
[0072] A“heteroaralkyl” or“heteroarylalkyl” is heteroaryl group connected, as a substituent, via an alkylene group. Examples include but are not limited to 2-thienylmethyl, 3-thienylmethyl, furylmethyl, thienylethyl, pyrrolylalkyl, pyridylalkyl, isoxazollylalkyl, and imidazolylalkyl. In some cases, the alkylene group is a lower alkylene group (i.e., a Ci-4 alkylene group).
[0073] As used herein,“carbocyclyl” means a non-aromatic cyclic ring or ring system containing only carbon atoms in the ring system backbone. When the carbocyclyl is a ring system, two or more rings may be joined together in a fused, bridged or spiro- connected fashion. Carbocyclyls may have any degree of saturation provided that at least one ring in a ring system is not aromatic. Thus, carbocyclyls include cycloalkyls, cycloalkenyls, and cycloalkynyls. The carbocyclyl group may have 3 to 20 carbon atoms, although the present definition also covers the occurrence of the term“carbocyclyl” where no numerical range is designated. The carbocyclyl group may also be a medium size carbocyclyl having 3 to 10 carbon atoms. The carbocyclyl group could also be a carbocyclyl having 3 to 6 carbon atoms. The carbocyclyl group may be designated as “C3-6 carbocyclyl” or similar designations. Examples of carbocyclyl rings include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, 2,3-dihydro-indene, bicycle[2.2.2]octanyl, adamantyl, and spiro[4.4]nonanyl.
[0074] A“(carbocyclyljalkyl” is a carbocyclyl group connected, as a substituent, via an alkylene group, such as "C4-10 (carbocyclyl)alkyl” and the like, including but not limited to, cyclopropylmethyl, cyclobutylmethyl, cyclopropylethyl, cyclopropylbutyl, cyclobutylethyl, cyclopropylisopropyl, cyclopentylmethyl, cyclopentylethyl, cyclohexylmethyl, cyclohexylethyl, cycloheptylmethyl, and the like. In some cases, the alkylene group is a lower alkylene group.
[0075] As used herein,“cycloalkyl” means a fully saturated carbocyclyl ring or ring system. Examples include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
[0076] As used herein,“cycloalkenyl” means a carbocyclyl ring or ring system having at least one double bond, wherein no ring in the ring system is aromatic. An example is cyclohexenyl.
[0077] As used herein,“heterocyclyl” means a non-aromatic cyclic ring or ring system containing at least one heteroatom in the ring backbone. Heterocyclyls may be joined together in a fused, bridged or spiro-connected fashion. Heterocyclyls may have any degree of saturation provided that at least one ring in the ring system is not aromatic. The heteroatom(s) may be present in either a non-aromatic or aromatic ring in the ring system. The heterocyclyl group may have 3 to 20 ring members (i.e., the number of atoms making up the ring backbone, including carbon atoms and heteroatoms), although the present definition also covers the occurrence of the term “heterocyclyl” where no numerical range is designated. The heterocyclyl group may also be a medium size heterocyclyl having 3 to 10 ring members. The heterocyclyl group could also be a heterocyclyl having 3 to 6 ring members. The heterocyclyl group may be designated as“3-6 membered heterocyclyl” or similar designations. In some embodiments, of the ring members, from 1 to 5 are heteroatoms, for example, the heterocyclyl may contain from 1 to 4 heteroatoms, from 1 to 3 heteroatoms, two heteroatoms, or one heteroatom. In preferred six membered monocyclic heterocyclyls, the heteroatom(s) are selected from one up to three of O, N or S, and in preferred five membered monocyclic heterocyclyls, the heteroatom(s) are selected from one or two heteroatoms selected from O, N, or S. Examples of heterocyclyl rings include, but are not limited to, azepinyl, acridinyl, carbazolyl, cinnolinyl, dioxolanyl, imidazolinyl, imidazolidinyl, morpholinyl, oxiranyl, oxepanyl, thiepanyl, piperidinyl, piperazinyl, dioxopiperazinyl, pyrrolidinyl, pyrrolidonyl, pyrrolidionyl, 4-piperidonyl, pyrazolinyl, pyrazolidinyl, 1,3-dioxinyl, 1,3-dioxanyl, 1,4-dioxinyl, 1,4-dioxanyl, 1,3-oxathianyl, 1,4- oxathiinyl, 1,4-oxathianyl, 2H--,2-oxazinyl, trioxanyl, hexahydro-l,3,5-triazinyl, 1,3- dioxolyl, 1,3 -dioxolanyl, 1,3-dithiolyl, 1,3-dithiolanyl, isoxazolinyl, isoxazolidinyl, oxazolinyl, oxazolidinyl, oxazolidinonyl, thiazolinyl, thiazolidinyl, 1 ,3-oxathiolanyl, indolinyl, isoindolinyl, tetrahydrofiiranyl, tetrahydropyranyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, tetrahydro-l,4-thiazinyl, thiamorpholinyl, dihydrobenzofuranyl, benzimidazolidinyl, and tetrahydroquinoline.
[0078] A “(heterocyclyl)alkyl” is a heterocyclyl group connected, as a substituent, via an alkylene group. Examples include, but are not limited to, imidazolinylmethyl and indolinylethyl.
[0079] As used herein,“acyl” refers to -C(=0)R, wherein R is selected from hydrogen, optionally substituted Ci-6 alkyl, halogen, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C6-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein. Non-limiting examples include formyl, acetyl, propanoyl, benzoyl, and acryl. [0080] An“O-carboxy" group refers to a“-0C(=O)R” group in which R is selected from hydrogen, optionally substituted Ci-6 alkyl, halogen, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C6-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
[0081] A“C-carboxy” group refers to a“-C(=0)OR” group in which R is selected from hydrogen, halogen, optionally substituted Ci-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C6-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein. A non-limiting example includes carboxyl (i.e., -C(=0)OH).
[0082] A“cyano” group refers to a“-CN” group.
[0083] A“cyanato” group refers to an“-OCN” group.
[0084] An“isocyanato” group refers to a“-NCO” group.
[0085] A“thiocyanato” group refers to a“-SCN” group.
[0086] An“isothiocyanato” group refers to an“-NCS” group.
[0087] A“sulfinyl” group refers to an“-S(=0)R” group in which R is selected from hydrogen, optionally substituted Ci-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted C2-0 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C6-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
[0088] A“sulfonyl” group refers to an“-SO2R” group in which R is selected from hydrogen, optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C6-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
[0089] An“S-sulfonamido” group refers to a“-SC>2NRARB” group in which RA and RB are each independently selected from hydrogen, halogen, optionally substituted Ci-6 alkyl, optionally substituted Ci-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C6-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
[0090] An“N-sulfonamido” group refers to a“-N(RA)SC>2RB" group in which RA and RB are each independently selected from hydrogen, halogen, optionally substituted Ci-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C6-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
[0091] A“C-amido” group refers to a“-C(=0)NRARB” group in which RA and RB are each independently selected from hydrogen, halogen, optionally substituted Ci-6 alkyl, optionally substituted Ci-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C6-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
[0092] An“N-amido” group refers to a“-N(RA)C(=0)RB” group in which RA and RB are each independently selected from hydrogen, halogen, optionally substituted C1-6 alkyl, optionally substituted Ci-6 alkoxy, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted Gs-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl, as defined herein.
[0093] An“O-carbamyl” group refers to a“-0C(=0)NRARB” group in which RA and RB are each independently selected from hydrogen, Ci-6 alkyl, Ci-6 alkenyl, C2-6 alkynyl, C3-7 carbocyclyl, C6-io aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein.
[0094] An“N-carbamyl” group refers to an“-N(RA)OC(=0)RB" group in which RA and RB are each independently selected from hydrogen, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 carbocyclyl, C6-io aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein.
[0095] An“O-thiocarbamyl” group refers to a“-OC(=S)NRARB” group in which RA and RB are each independently selected from hydrogen, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 carbocyclyl, a C6-io aryl, 5-10 membered heteroaryl, and 5-10 membered heterocyclyl, as defined herein.
[0096] An“N-thiocarbamyl” group refers to an “-N(RA)OC(=S)RB” group in which RA and RB are each independently selected from hydrogen, Ci-6 alkyl, C2-6 alkenyl, C2- 6 alkynyl, C3-7 carbocyclyl, C6-io aryl, 5-10 member ed heteroaryl, and 5-10 member ed heterocyclyl, as defined herein.
[0097] An“amino” group refers to a“-NRARB” group in which RA and RB are each independently selected from hydrogen, halogen, optionally substituted C1-6 alkyl, optionally substituted C2-6 alkenyl, optionally substituted C2-6 alkynyl, optionally substituted C3-7 carbocyclyl, optionally substituted C6-io aryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl as defined herein. A nonlimiting example includes free amino (i.e., -NH2).
[0098] An “aminoalkyl” group refers to an amino group connected via an alkylene group.
[0099] An“alkoxyalkyl” group refers to an alkoxy group connected via an alkylene group, such as a“C2-8 alkoxyalkyl” and the like.
[0100] As used herein, a substituted group is derived from the unsubstituted parent group in which there has been an exchange of one or more hydrogen atoms for another atom or group. Unless otherwise indicated, when a group is deemed to be“substituted,” it is meant that the group is substituted with one or more substituents independently selected from C1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkynyl, C1-C6 heteroalkyl, C3-C7 carbocyclyl (optionally substituted with halo, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, and C1-C6 haloalkoxy), C3- C7-carbocyclyl-Ci-C6-alkyl (optionally substituted with halo, C1-C6 alkyl, C1-C6 alkoxy, Ci-C6 haloalkyl, and C1-C6 haloalkoxy), 3-10 membered heterocyclyl (optionally substituted with halo, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, and C1-C6 haloalkoxy), 3-10 membered heterocyclyl-C1-C6-alkyl (optionally substituted with halo, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, and C1-C6 haloalkoxy), aiyl (optionally substituted with halo, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, and C1-C6 haloalkoxy), aryl(C1-C6)alkyl (optionally substituted with halo, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, and C1-C6 haloalkoxy), 5- 10 membered heteroaryl (optionally substituted with halo, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, and C1-C6 haloalkoxy), 5-10 membered heteroaryl( C1-C6)alkyl (optionally substituted with halo, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, and C1-C6 haloalkoxy), halo, cyano, hydroxy, C1-C6 alkoxy, C1-C6 alkoxy(Ci-C6)alkyl (i.e., ether), aryloxy, sulfhydryl (mercapto), halo(C1-C6)alkyl (e.g., -CF3), halo(Ci-C6)alkoxy (e.g., -OCF3), C1-C6 alkylthio, arylthio, amino, amino(Ci-C6)alkyl, nitro, O-carbamyl, N-carbamyl, O- thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C- carboxy, O-carboxy, acyl, cyanato, isocyanato, thiocyanate, isothiocyanato, sulfinyl, sulfonyl, and oxo (=0). Wherever a group is described as“optionally substituted” that group can be substituted with the above substituents.
[0101] It is to be understood that certain radical naming conventions can include either a mono-radical or a di-radical, depending on the context. For example, where a substituent requires two points of attachment to the rest of the molecule, it is understood that the substituent is a di-radical. For example, a substituent identified as alkyl that requires two points of attachment includes di-radicals such as -CH2-, -CH2CH2-, -CH2CH(CH3)CH2-, and the like. Other radical naming conventions clearly indicate that the radical is a di-radical such as“alkylene” or“alkenylene.”
[0102] When two R groups are said to form a ring (e.g., a carbocyclyl, heterocyclyl, aryl, or heteroaryl ring)“together with the atom to which they are attached,” it is meant that the collective unit of the atom and the two R groups are the recited ring. The ring is not otherwise limited by the definition of each R group when taken individually. For example, when the following substructure is present:
Figure imgf000029_0001
and R1 and R2 are defined as selected from the group consisting of hydrogen and alkyl, or R1 and R2 together with the nitrogen to which they are attached form a heterocyclyl, it is meant that R1 and R2 can be selected from hydrogen or alkyl, or alternatively, the substructure has structure:
Figure imgf000029_0002
where ring A is a heterocyclyl ring containing the depicted nitrogen. [0103] Similarly, when two“adjacent” R groups are said to form a ring“together with the atom to which they are attached,” it is meant that the collective unit of the atoms, intervening bonds, and the two R groups are the recited ring. For example, when the following substructure is present:
Figure imgf000030_0001
and R1 and R2 are defined as selected from the group consisting of hydrogen and alkyl, or R1 and R2 together with the atoms to which they are attached form an aryl or carbocyclyl, it is meant that R1 and R2 can be selected from hydrogen or alkyl, or alternatively, the substructure has structure:
Figure imgf000030_0002
where A is an aryl ring or a carbocyclyl containing the depicted double bond.
[0104] Wherever a substituent is depicted as a di-radical (i.e., has two points of attachment to the rest of the molecule), it is to be understood that the substituent can be attached in any directional configuration unless otherwise indicated. Thus, for example, a substituent depicted as -AE- or includes the substituent being oriented such
Figure imgf000030_0003
that the A is attached at the leftmost attachment point of the molecule as well as the case in which A is attached at the rightmost attachment point of the molecule.
[0105] “Subject” as used herein, means a human or a non-human mammal, e.g., a dog, a cat, a mouse, a rat, a cow, a sheep, a pig, a goat, a non-human primate or a bird, e.g., a chicken, as well as any other vertebrate or invertebrate.
[0106] The term“mammal” is used in its usual biological sense. Thus, it specifically includes, but is not limited to, primates, including simians (chimpanzees, apes, monkeys) and humans, cattle, horses, sheep, goats, swine, rabbits, dogs, cats, rodents, rats, mice guinea pigs, or the like. [0107] The term “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. In addition, various adjuvants such as are commonly used in the art may be included. Considerations for the inclusion of various components in pharmaceutical compositions are described, e.g., in Gilman et al. (Eds.) (1990); Goodman and Gilman’s: The Pharmacological Basis of Therapeutics, 8th Ed., Pergamon Press.
[0108] A therapeutic effect relieves, to some extent, one or more of the symptoms of a disease or condition, and includes curing a disease or condition. “Curing” means that the symptoms of a disease or condition are eliminated; however, certain long-term or permanent effects may exist even after a cure is obtained (such as extensive tissue damage).
[0109] “Treat,”“treatment,” or“treating,” as used herein refers to administering a compound or pharmaceutical composition to a subject for prophylactic and/or therapeutic purposes. The term“prophylactic treatment” refers to treating a subject who does not yet exhibit symptoms of a disease or condition, but who is susceptible to, or otherwise at risk of, a particular disease or condition, whereby the treatment reduces the likelihood that the patient will develop the disease or condition. The term “therapeutic treatment” refers to administering treatment to a subject already suffering from a disease or condition.
[0110] “Metabolites” of the compounds disclosed herein include active species that are produced upon introduction of the compounds into the biological milieu.
[0111] “Solvate” refers to the compound formed by the interaction of a solvent and a compound described herein, a metabolite, or salt thereof. Suitable solvates are pharmaceutically acceptable solvates including hydrates.
[0112] The term“pharmaceutically acceptable salt” refers to salts that retain the biological effectiveness and properties of a compound, which are not biologically or otherwise undesirable for use in a pharmaceutical. In many cases, the compounds herein are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic 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. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like; particularly preferred are the ammonium, potassium, sodium, calcium and magnesium salts. Organic bases from which salts can be derived include, for example, primaiy, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. Many such salts are known in the art, as described in WO 87/05297, Johnston et al., published September 11, 1987 (incorporated by reference herein in its entirety).
Methods of Preparation
[0113] The compounds disclosed herein may be synthesized by methods described below, or by modification of these methods. Ways of modifying the methodology include, among others, temperature, solvent, reagents, etc., known to those skilled in the art. In general, during any of the processes for preparation of the compounds disclosed herein, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry (ed. J.F.W. McOmie, Plenum Press, 1973); and P.G.M Green, T.W. Wutts, Protecting Groups in Organic Synthesis (3rd ed.) Wiley, New York (1999), which are both hereby incorporated herein by reference in their entirety The protecting groups may be removed at a convenient subsequent stage using methods known from the art. Synthetic chemistry transformations useful in synthesizing applicable compounds are known in the art and include, e.g., those described in R Larock, Comprehensive Organic Transformations, VCH Publishers, 1989, or L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons, 1995, which are both hereby incorporated herein by reference in their entirety. The routes shown and described herein are illustrative only and are not intended, nor are they to be construed, to limit the scope of the claims in any manner whatsoever. Those skilled in the art will be able to recognize modifications of the disclosed syntheses and to devise alternate routes based on the disclosures herein; all such modifications and alternate routes are within the scope of the claims.
[0114] Scheme I is provided for the guidance of the reader, and represents an example of a general strategy for making the compounds described herein. Other methods for preparing the compounds described herein will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples.
[0115] Compounds of structure 4 can be prepared from precursors of structure 1 by protection of the diol, reduction of the carboxyl to the corresponding aldehyde (structure 3), acetalation, and then deprotection. Compounds of structure 5 can be obtained by acylation or further acetalation of the compounds of structure 4.
[0116] Compounds of structure 6 can be prepared from precursors of structure 1 by esterification followed by acetalation. Compounds of structure 7 can be prepared by direct acetalation of precursors of structure 1. Compounds of structure 8 can be obtained from precursors of structure 1 by lactonization followed by acetalation.
[0117] Compounds of structure 9 can be obtained by direct reduction of precursors of structure 1. Compounds of structures 10 and 11 can be prepared from compounds of structure 9 by treatment with formaldehyde.
Scheme I
Figure imgf000034_0001
Administration and Pharmaceutical Compositions
[0118] The compounds are administered at a therapeutically effective dosage. While human dosage levels have yet to be optimized for the compounds described herein, generally, a daily dose may be from about 0.25 mg/kg to about 120 mg/kg or more of body weight, from about 0.5 mg/kg or less to about 70 mg/kg, from about 1.0 mg/kg to about 50 mg/kg of body weight, or from about 1.5 mg/kg to about 10 mg/kg of body weight Thus, for administration to a 70 kg person, the dosage range would be from about 17 mg per day to about 8000 mg per day, from about 35 mg per day or less to about 7000 mg per day or more, from about 70 mg per day to about 6000 mg per day, from about 100 mg per day to about 5000 mg per day, or from about 200 mg to about 3000 mg per day. The amount of active compound administered will, of course, be dependent on the subject and disease state being treated, the severity of the affliction, the manner and schedule of administration and the judgment of the prescribing physician.
[0119] Administration of the compounds disclosed herein or the pharmaceutically acceptable salts thereof can be via any of the accepted modes of administration for agents that serve similar utilities including, but not limited to, orally, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarilly, vaginally, rectally, or intraocularly. Oral and parenteral administrations are customary in treating the indications that are the subject of the preferred embodiments.
[0120] The compounds described above can be formulated into pharmaceutical compositions for use in treatment of these conditions. Standard pharmaceutical formulation techniques are used, such as those disclosed in Remington's The Science and Practice of Pharmacy, 21st Ed., Lippincott Williams & Wilkins (2005), incorporated by reference in its entirety. Accordingly, some embodiments include pharmaceutical compositions comprising: (a) a safe and therapeutically effective amount of a compound described herein (including enantiomers, diastereoisomers, tautomers, polymorphs, and solvates thereof), or pharmaceutically acceptable salts thereof; and (b) a pharmaceutically acceptable carrier, diluent, excipient or combination thereof.
[0121] In addition to the compounds described above, some embodiments include compositions containing a pharmaceutically-acceptable carrier. The term“pharmaceutically acceptable carrier” or“pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. In addition, various adjuvants such as are commonly used in the art may be included. Considerations for the inclusion of various components in pharmaceutical compositions are described, e.g., in Gilman et al. (Eds.) (1990); Goodman and Gilman’s: The Pharmacological Basis of Therapeutics, 8th Ed., Pergamon Press, which is incorporated herein by reference in its entirety.
[0122] Some examples of substances, which can serve as pharmaceutically- acceptable carriers or components thereof, are sugars, such as lactose, glucose and sucrose; starches, such as com starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and methyl cellulose; powdered tragacanth; malt; gelatin; talc; solid lubricants, such as stearic acid and magnesium stearate; calcium sulfate; vegetable oils, such as peanut oil, cottonseed oil, sesame oil, olive oil, com oil and oil of theobroma; polyols such as propylene glycol, glycerine, sorbitol, mannitol, and polyethylene glycol; alginic acid; emulsifiers, such as the TWEENS; wetting agents, such sodium lauryl sulfate; coloring agents; flavoring agents; tableting agents, stabilizers; antioxidants; preservatives; pyrogen-free water; isotonic saline; and phosphate buffer solutions.
[0123] The choice of a pharmaceutically-acceptable carrier to be used in conjunction with the subject compound is basically determined by the way the compound is to be administered.
[0124] The compositions described herein are preferably provided in unit dosage form. As used herein, a "unit dosage form" is a composition containing an amount of a compound that is suitable for administration to an animal, preferably mammal subject, in a single dose, according to good medical practice. The preparation of a single or unit dosage form however, does not imply that the dosage form is administered once per day or once per course of therapy. Such dosage forms are contemplated to be administered once, twice, thrice or more per day and may be administered as infusion over a period of time (e.g., from about 30 minutes to about 2-6 hours), or administered as a continuous infusion, and may be given more than once during a course of therapy, though a single administration is not specifically excluded. The skilled artisan will recognize that the formulation does not specifically contemplate the entire course of therapy and such decisions are left for those skilled in the art of treatment rather than formulation.
[0125] The compositions described above may be in any of a variety of suitable forms for a variety of routes for administration, for example, for oral, nasal, rectal, topical (including transdermal), ocular, intracerebral, intracranial, intrathecal, intra-arterial, intravenous, intramuscular, or other parental routes of administration. The skilled artisan will appreciate that oral and nasal compositions comprise compositions that are administered by inhalation, and made using available methodologies. Depending upon the particular route of administration desired, a variety of pharmaceutically-acceptable carriers well-known in the art may be used. Pharmaceutically-acceptable carriers include, for example, solid or liquid fillers, diluents, hydrotropies, surface-active agents, and encapsulating substances. Optional pharmaceutically-active materials may be included, which do not substantially interfere with the inhibitory activity of the compound. The amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound. Techniques and compositions for making dosage forms useful in the methods described herein are described in the following references, all incorporated by reference herein: Modem Pharmaceutics, 4th Ed., Chapters 9 and 10 (Banker & Rhodes, editors, 2002); Lieberman et al, Pharmaceutical Dosage Forms: Tablets (1989); and Ansel, Introduction to Pharmaceutical Dosage Forms 8th Edition (2004).
[0126] Various oral dosage forms can be used, including such solid forms as tablets, capsules, granules and bulk powders. Tablets can be compressed, tablet triturates, enteric-coated, sugar-coated, film-coated, or multiple-compressed, containing suitable binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, flow- inducing agents, and melting agents. Liquid oral dosage forms include aqueous solutions, emulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules, and effervescent preparations reconstituted from effervescent granules, containing suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, melting agents, coloring agents and flavoring agents.
[0127] The pharmaceutically-acceptable carrier suitable for the preparation of unit dosage forms for peroral administration is well-known in the art. Tablets typically comprise conventional pharmaceutically-compatible adjuvants as inert diluents, such as calcium carbonate, sodium carbonate, mannitol, lactose and cellulose; binders such as starch, gelatin and sucrose; disintegrants such as starch, alginic acid and croscarmelose; lubricants such as magnesium stearate, stearic acid and talc. Glidants such as silicon dioxide can be used to improve flow characteristics of the powder mixture. Coloring agents, such as the FD&C dyes, can be added for appearance. Sweeteners and flavoring agents, such as aspartame, saccharin, menthol, peppermint, and fruit flavors, are useful adjuvants for chewable tablets. Capsules typically comprise one or more solid diluents disclosed above. The selection of carrier components depends on secondary considerations like taste, cost, and shelf stability, which are not critical, and can be readily made by a person skilled in the art. [0128] Peroral compositions also include liquid solutions, emulsions, suspensions, and the like. The pharmaceutically-acceptable carriers suitable for preparation of such compositions are well known in the art. Typical components of carriers for syrups, elixirs, emulsions and suspensions include ethanol, glycerol, propylene glycol, polyethylene glycol, liquid sucrose, sorbitol and water. For a suspension, typical suspending agents include methyl cellulose, sodium carboxymethyl cellulose, AVICEL RC-591, tragacanth and sodium alginate; typical wetting agents include lecithin and polysorbate 80; and typical preservatives include methyl paraben and sodium benzoate. Peroral liquid compositions may also contain one or more components such as sweeteners, flavoring agents and colorants disclosed above.
[0129] Such compositions may also be coated by conventional methods, typically with pH or time-dependent coatings, such that the subject compound is released in the gastrointestinal tract in the vicinity of the desired topical application, or at various times to extend the desired action. Such dosage forms typically include, but are not limited to, one or more of cellulose acetate phthalate, polyvinylacetate phthalate, hydroxypropyl methyl cellulose phthalate, ethyl cellulose, Eudragit coatings, waxes and shellac.
[0130] Compositions described herein may optionally include other drug actives.
[0131] Other compositions useful for attaining systemic delivery of the subject compounds include sublingual, buccal and nasal dosage forms. Such compositions typically comprise one or more of soluble filler substances such as sucrose, sorbitol and mannitol; and binders such as acacia, microcrystalline cellulose, carboxymethyl cellulose and hydroxypropyl methyl cellulose. Glidants, lubricants, sweeteners, colorants, antioxidants and flavoring agents disclosed above may also be included.
[0132] A liquid composition, which is formulated for topical ophthalmic use, is formulated such that it can be administered topically to the eye. The comfort should be maximized as much as possible, although sometimes formulation considerations (e.g. drug stability) may necessitate less than optimal comfort. In the case that comfort cannot be maximized, the liquid should be formulated such that the liquid is tolerable to the patient for topical ophthalmic use. Additionally, an ophthalmically acceptable liquid should either be packaged for single use, or contain a preservative to prevent contamination over multiple uses. [0133] For ophthalmic application, solutions or medicaments are often prepared using a physiological saline solution as a major vehicle. Ophthalmic solutions should preferably be maintained at a comfortable pH with an appropriate buffer system. The formulations may also contain conventional, pharmaceutically acceptable preservatives, stabilizers and surfactants.
[0134] Preservatives that may be used in the pharmaceutical compositions disclosed herein include, but are not limited to, benzalkonium chloride, PHMB, chlorobutanol, thimerosal, phenylmercuric, acetate and phenylmercuric nitrate. A useful surfactant is, for example, Tween 80. Likewise, various useful vehicles may be used in the ophthalmic preparations disclosed herein. These vehicles include, but are not limited to, polyvinyl alcohol, povidone, hydroxypropyl methyl cellulose, poloxamers, carboxymethyl cellulose, hydroxyethyl cellulose and purified water.
[0135] Tonicity adjustors may be added as needed or convenient They include, but are not limited to, salts, particularly sodium chloride, potassium chloride, mannitol and glycerin, or any other suitable ophthalmically acceptable tonicity adjustor.
[0136] Various buffers and means for adjusting pH may be used so long as the resulting preparation is ophthalmically acceptable. For many compositions, the pH will be between 4 and 9. Accordingly, buffers include acetate buffers, citrate buffers, phosphate buffers and borate buffers. Acids or bases may be used to adjust the pH of these formulations as needed.
[0137] In a similar vein, an ophthalmically acceptable antioxidant includes, but is not limited to, sodium metabisulfite, sodium thiosulfate, acetylcysteine, butylated hydroxyanisole and butylated hydroxytoluene.
[0138] Other excipient components, which may be included in the ophthalmic preparations, are chelating agents. A useful chelating agent is edetate disodium, although other chelating agents may also be used in place or in conjunction with it
[0139] For topical use, creams, ointments, gels, solutions or suspensions, etc., containing the compound disclosed herein are employed. Topical formulations may generally be comprised of a pharmaceutical carrier, co-solvent, emulsifier, penetration enhancer, preservative system, and emollient [0140] For intravenous administration, the compounds and compositions described herein may be dissolved or dispersed in a pharmaceutically acceptable diluent, such as a saline or dextrose solution. Suitable excipients may be included to achieve the desired pH, including but not limited to NaOH, sodium carbonate, sodium acetate, HC1, and citric acid. In various embodiments, the pH of the final composition ranges from 2 to 8, or preferably from 4 to 7. Antioxidant excipients may include sodium bisulfite, acetone sodium bisulfite, sodium formaldehyde, sulfoxylate, thiourea, and EDTA. Other non-limiting examples of suitable excipients found in the final intravenous composition may include sodium or potassium phosphates, citric acid, tartaric acid, gelatin, and carbohydrates such as dextrose, mannitol, and dextran. Further acceptable excipients are described in Powell, et al., Compendium of Excipients for Parenteral Formulations, PDA J Pharm Sci and Tech 1998, 52 238-311 and Nema et al., Excipients and Their Role in Approved Injectable Products: Current Usage and Future Directions, PDA J Pharm Sci and Tech 2011, 65 287-332, both of which are incorporated herein by reference in their entirety. Antimicrobial agents may also be included to achieve a bacteriostatic or fungistatic solution, including but not limited to phenylmercuric nitrate, thimerosal, benzethonium chloride, benzalkonium chloride, phenol, cresol, and chlorobutanol.
[0141] The compositions for intravenous administration may be provided to caregivers in the form of one more solids that are reconstituted with a suitable diluent such as sterile water, saline or dextrose in water shortly prior to administration In other embodiments, the compositions are provided in solution ready to administer parenterally. In still other embodiments, the compositions are provided in a solution that is further diluted prior to administration. In embodiments that include administering a combination of a compound described herein and another agent, the combination may be provided to caregivers as a mixture, or the caregivers may mix the two agents prior to administration, or the two agents may be administered separately.
[0142] The actual dose of the active compounds described herein depends on the specific compound, and on the condition to be treated; the selection of the appropriate dose is well within the knowledge of the skilled artisan.
Methods of Treatment [0143] Some embodiments of the present invention include methods of treating a liver disease, disorder, or condition by administering the compounds or pharmaceutical compositions described herein to a subject in need thereof. In some such embodiments, the liver disease, disorder, or condition involves the metabolism, storage, or homeostatic control of biochemical end products such as cholesterol, fatty acids, bile, triglycerides, plasma proteins, carrier proteins, lipoproteins, acute phase proteins, apolipoproteins, carbohydrates, hemostatic factors, fibrinolysis factors, thrombopoietin, insulin-like growth factor 1, hepcidin, angiotensinogen, vitamin A, vitamin D, vitamin B12, vitamin K, iron, copper, or catalase. In some particular embodiments, the liver disease, disorder, or condition being treated is liver fibrosis, cirrhosis, hepatitis, alcoholic liver disease, fatty liver, liver cancer, hemochromatosis, Wilson’s disease, primary sclerosing cholangitis, alpha 1-antitrypsin deficiency, hyperglycemia, hypoglycemia, or diabetes.
[0144] Some embodiments of the present invention include methods of treating a cardiovascular disease, disorder, or condition, by administering the compounds or pharmaceutical compositions described herein to a subject in need thereof. In some particular embodiments, the cardiovascular disease, disorder, or condition is dyslipidemia or atherosclerosis. In other particular embodiments, the cardiovascular disease, disorder, or condition is hyperlipidemia.
[0145] Some embodiments of the present invention include methods of altering blood lipid concentration by administering the compounds or pharmaceutical compositions described herein to a subject in need thereof.
[0146] Some embodiments of the present invention include methods of inhibiting HMG-CoA reductase by administering the compounds or pharmaceutical compositions described herein to a subject in need thereof.
[0147] Some embodiments of the present invention include methods of reducing the side effects of liver-targeting or liver-activated drugs by administering the compounds or pharmaceutical compositions described herein, rather than their corresponding active drag compound, to a subject in need thereof. In some such embodiments, the corresponding active drug compound is a statin. Examples side effects that can be reduced include headache, difficulty sleeping, flushing of the skin, muscle aches, muscle tenderness, muscle weakness, drowsiness, dizziness, nausea, vomiting, abdominal pain, bloating, gas, diarrhea, constipation, or rash.
[0148] Some embodiments of the present invention include methods of increasing the therapeutic index of liver-targeting or liver-activated therapeutic agents.
[0149] In some embodiments, the subject is a mammal.
[0150] In some embodiments, the subject is a human.
[0151] Further embodiments include administering a combination of compounds to a subject in need thereof. A combination can include a compound or pharmaceutical composition described herein with an additional medicament.
[0152] Some embodiments include co-administering a compound or pharmaceutical composition described herein, with an additional medicament. By“co- administration,” it is meant that the two or more agents may be found in the patient’s bloodstream at the same time, regardless of when or how they are actually administered. In one embodiment, the agents are administered simultaneously. In one such embodiment, administration in combination is accomplished by combining the agents in a single dosage form. In another embodiment, the agents are administered sequentially. In one embodiment, the agents are administered through the same route, such as orally. In another embodiment, the agents are administered through different routes, such as one being administered orally and another being administered intravenously.
[0153] In some embodiments, the additional medicament is a cholesterol absorption inhibitor, a hypertension medicament, niacin, or aspirin. In some such embodiments, the cholesterol absorption inhibitor is ezetimibe. In some such embodiments, the hypertension medicament is a diuretic, a beta-blocker, an angiotensin-converting enzyme inhibitor, an angiotensin receptor blocker, a calcium channel blocker, an alpha blocker, an alpha-2 receptor agonist, a combined alpha and beta-blocker, a central agonist, a peripheral adrenergic inhibitor, or a vasodilator. Examples of hypertension medicaments that can be used as the additional medicament include acebutolol, alpha methyldopa, amiloride hydrochloride, amlodipine besylate, atenolol, benazepril hydrochloride, betaxolol, bepridil, bisoprolol fumarate, bumetanide, candesartan, captopril, carteolol hydrochloride, carvedilol, chlorthalidone, chlorothiazide, clonidine hydrochloride, diltiazem hydrochloride, doxazosin mesylate, enalapril maleate, eprosartan mesylate, felodipine, fosinopril sodium, furosemide, hydralazine hydrochloride, hydrochlorothiazide, guanabenz acetate, guanadrel, guanethidine monosulfate, guanfacine hydrochloride, indapamide, irbesarten, isradipine, labetalol hydrochloride, lisinopril, losartan potassium, methyldopa, metolazone, metoprolol succinate, metoprolol tartrate, minoxidil, moexipril, nadolol, nicardipine, nifedipine, nisoldipine, penbutolol sulfate, perindopril, pindolol, prazosin hydrochloride, propranolol hydrochloride, quinapril hydrochloride, ramipril, reserpine, telmisartan, terazosin hydrochloride, trandolapril, triamterene, solotol hydrochloride, spironolactone, timolol maleate, valsartan, and verapamil hydrochloride.
[0154] To further illustrate this invention, the following examples are included. The examples should not, of course, be construed as specifically limiting the invention. Variations of these examples within the scope of the claims are within the purview of one skilled in the art and are considered to fall within the scope of the invention as described, and claimed herein. The reader will recognize that the skilled artisan, armed with the present disclosure, and skill in the art is able to prepare and use the invention without exhaustive examples. The following examples will further describe the present invention, and are used for the purposes of illustration only, and should not be considered as limiting.
EXAMPLES
[0155] Some compounds of Formula I, la, lb, Ic, Id, Ie, If, Ig, and P can be prepared as described below.
Example 1
[0156] N-( 5-(Y3 S 5R.EV6-11 3 -Dioxolan-2-vll-3- 5-dihvdroxvhex- 1 -en- 1 -vl V4-/4- fluoroDhenvlV6-isoDropvlpvrimidin-2-vlVN-methvlmethanesulfonamide (Compound 1011
Figure imgf000043_0001
[0157] To a solution of rosuvastatin (10.0 g, 20.8 mmol) in DMF (75.0 mL) was added 3-bromoprop-l-ene (4.20 g, 34.7 mmol, 1.67 eq). The solution was stirred at 25°C and monitored by TLC. After 16 hours, the reaction mixture was concentrated under reduced pressure, diluted with H2O, and extracted with DCM After drying over NazSCU, the combined organic layers were concentrated under reduced pressure. The concentrate was purified by column chromatography (PE/EA = 1:1) to give rosuvastatin allyl ester (6.41 g, 12.3 mmol, 59% yield) as a light-yellow oil.
[0158] Imidazole (5.56 g, 81.7 mmol, 6.00 eq) was added to a solution of the allyl ester (6.91 g, 13.6 mmol) in DMF (50 mL), and then a solution of TIPDSC1 (6.33 g, 27.22 mmol, 2.00 eq) in DMF (25 mL) was added dropwise. The resulting mixture was stirred at 25°C for 16 hours and monitored by TLC. Afterward, the reaction mixture was concentrated under reduced pressure, diluted with H2O, and extracted with DCM The combined organic layers were dried over NaaSCU and concentrated to give the crude mixture. Purification by column chromatography (PE/EA = 5:1) afforded TIPD S -protected rosuvastatin allyl ester (10.0 g, 13.1 mmol, 96% yield) as a light-yellow oil.
[0159] To a solution of the TIPDS-protected rosuvastatin allyl ester (12.9 g, 16.8 mmol) in EtOH (200 mL) was added NaBHt (1.27 g, 33.7 mmol, 2.00 eq). The resulting mixture was stirred at 40°C and monitored by TLC. After 16 hours, the reaction mixture was filtered, washed with EtOH, and concentrated under reduced pressure. Purification by column chromatography (PE: EA = 5:1) yielded the primary alcohol (6.56 g, 9.24 mmol, 55% yield) as a light-yellow oil.
[0160] To a solution of the primary alcohol (6.38 g, 8.99 mmol) in DCM (120 mL) was added Dess-Martin periodinane (4.58 g, 10.79 mmol, 1.20 eq). The reaction mixture was stirred at 25°C for 16 hours while monitoring by TLC. The reaction mixture was filtered, concentrated under reduced pressure, diluted with DCM, and then washed with water. After drying over NaaSO-r, the combined organic layers were concentrated under reduced pressure. The concentrate was purified by column chromatography (PE/EA = 10:1) to afford the aldehyde (4.17 g, 5.89 mmol, 65% yield).
[0161] Ethylene glycol (87.7 mg, 1.41 mmol, 2.00 eq), TosOH (60.8 mg, 0.353 mmol, 0.50 eq) and MgS04 (10 g, 83 mmol) were added to a solution of the aldehyde (500 mg, 0.706 mmol) in DCM (50 mL). The resulting mixture was stirred at 60°C for 48 hours and until HPLC (ET12043-33-P1B) indicated that 30% of the aldehyde was consumed. The reaction mixture was filtered and the filtrate washed with DCM Afterward, the combined organics were concentrated, diluted with DCM, and washed with water. The combined organic layers were dried over Na2SC>4 and concentrated trader reduced pressure. Purification by column chromatography (PE/EA = 25:1) gave the acetal (177 mg, 0.235 mmol, 33% yield) as a light-yellow oil.
[0162] To a solution of the acetal (602 mg, 0.80 mmol) in THE (50.0 mL) was added TEA-3HF (774 mg, 4.80 mmol, 6.00 eq). The resulting mixture was stirred at 30°C for 16 hours and until TLC indicated that most of the acetal was consumed. The reaction mixture was concentrated and purified by column chromatography (PE/EA = 25/1 to 3/1) to afford Compound 101 (328 mg, 0.64 mmol, 80% yield) as light-yellow oil. XH NMR (400 MHz, CDCh): 57.65-7.62 (m, 2H), 7.09-7.05 (m, 2H), 6.63-6.58 (dd, J= 16.0 and 1.65, 1H), 5.47-5.41 (dd, J = 16.0 and 5.07, 1H), 5.00-4.98 (t, 1H), 4.45-4.42 (m, 1H), 4.17-4.12 (m, 1H), 4.02-3.98 (m, 2H), 3.92-3.82 (m, 3H), 3.55 (s, 3H),3.50 (s, 3H), 3.40-3.31 (m, 1H), 1.89-1.81 (m, 2H), 1.59-1.50 (m, 3H), 1.44-1.40 (m, 1H), 1.24-1.25(d, J = 6.62, 6H). [M+H]+ calculated for C24H32FN3O6S: 510.21; found: 510.2.
Example 2
[0163] N-(5-(Y3 S.5R-EV6-11.3 -Dioxan-2-vll-3.5-dihvdroxvhex- 1 -en- 1 -nP-4-f 4- fluorophenvll-6-isopropvlpvrimidin-2-vlVN-methvlmethanesulfonamide (Compound 1021
Figure imgf000045_0002
[0164] Compound 102 was prepared according to Scheme I from rosuvastatin and 1 ,3-dihydroxypropane in a manner similar to that of Compound 101. [M+H]+ calculated for C25H34FN3O6S: 524.23; found: 524.3.
Example 3
[0165] N-(5-( CEY2-( ( 4S.6RV 6-( ( 1.3 -Dioxolan-2-vllmethvr)-2-oxo- 1.3-dioxan-4- vlWinvll-4- fluorophenvll-6-isopropvlpvrimidin-2-vll-N-methvlmethanesulfonamide
Figure imgf000045_0001
(Compound 1031
Figure imgf000046_0001
[0166] Compound 103 was prepared from treatment of Compound 101 with carbonyldiimidazole. [M+H]+ calculated for C25H30FN3O7S: 536.19; found: 536.1.
Example 4
[0167] ( 2R.4S.E)- 1 -P .3-Dioxan-2-vlV6-f , 4-( 4-fluorophenvl)-6-isopropvl-2-(N- methvlmethvlsulfonamido)pvrimidin-5-vl)-4-hvdroxvhex-5-en-2-vl isobutvrate (Compound
104) and (3S.5R.El-6-n.3-dioxan-2-vl)-l-(4-(4-fluorophenvlV6-isopropvl-2-(N- methvlmethvlsulfonamido)pvrimidin-5-vl)-5-hvdroxvhex-l-en-3-vl isobutvrate (Compound
105
Figure imgf000046_0002
[0168] Compounds 104 and 105 were prepared as a mixture from treatment of Compound 102 with isobutyryl chloride in the presence of pyridine. [M+H]+ calculated for C29H40FN3O7S: 594.27; found: 594.3.
Example 5
[0169] (2R.4S.E)-l-(1.3-Dioxan-2-vD-6-(4-(4-fluorophenvl)-6-isopropvl-2-(N- methvlmethvlsulfonamido)pvrimidin-5-vl)hex-5-ene-2.4-divl _ bisf 2-methvlpropanoate)
(Compound 1061
Figure imgf000047_0001
[0170] Compounds 106 were prepared from treatment of Compound 102 with isobutyryl chloride in the presence of pyridine. [M+H]+ calculated for C33H46FN3O8S: 664.31; found: 664.3.
Example 6
[0171] N-(5-((3 S.5R.EV6-( 4-Ethvl- 1.S-dioxolan^-vlVS.S-dihvdroxvhex- 1 -en- 1 - vl)-4-(4-fluoroohenvl)-6-isooropvlpvrimidin-2-vl)-N-methvlmethanesulfonamide
(Compound 107)
Figure imgf000047_0002
[0172] Compound 107 was prepared according to Scheme I from rosuvastatin and (±)-l,2-dihydroxybutane in a manner similar to that of Compound 101. [M+H]+ calculated for CaeHs&FNsOeS: 538.24; found: 538.2.
Example 7
[0173] N-(5-((3S.5R.EV6-((4S.6Sl-4.6-Dimethvl-1.3-dioxan-2-vn-3.5- dihvdroxvhex- 1 -en- 1 -vl)-4-(4-fluorophenvD-6-isopropvlpvrimidin-2-vlVN- methvlmethanesulfonamide (Compound 1081 and N-(5-((3S.5R.E)-6-((4R.6RV4.6-dimethvl-
1.3-dioxan-2-vfl-3.5-dihvdroxvhex-l-en-l-vD-4-(4-fluorophenvlV6-isopropvlpvrimidin-2- vll-N-methvlmethanesulfonamide (Compound 1091
Figure imgf000048_0001
[0174] Compounds 108 and 109 were prepared as a mixture according to Scheme I from rosuvastatin and (±)-2,4-dihydroxypentane in a manner similar to that of Compound 101. [M+H]+ calculated for C27H38FN3O6S: 552.26; found: 552.2.
Example 8
G01751 N-15-ff3S.5R.El-6-rr2S.4S.6Rl-4.6-Dimethvl-l.3-dioxan-2-vn-3.5- dihvdroxvhex- 1 -en- 1 -vl)-4-(4-fluorophenvl)-6-isopropvlpvrimidin-2-vl 1-N- methvlmethanesulfonamide (Compound 110) and N-(5-((3S.5R.EV6-((2R.4R.6Rl-4.6- dimethvl-1 3-dioxan-2-vlV3.5-dihvdroxvhex- 1 -en- 1 -vl)-4-(4-fluorophenvD-6- isopropvlpvrimidin-2-vll-N-methvlmethanesulfonamide ( Compound 111)
Figure imgf000048_0002
[0176] Compounds 110 and 111 were isolated as a mixture from the reaction in
Example 7 operated according to Scheme I from rosuvastatin and (±)-2,4-dihydroxypentane in a manner similar to that of Compound 101. [M+H]+ calculated for C27H38FN3O6S: 552.26; found: 552.2.
Example 9
[0177] N-(4-(4-FluoroDhenvll-6-isoDroDvl-5-((3S.5S.EV3.5.7-trihvdroxvhept-l- en-l-vllpvrimidin-2-vlVN-methvlmethanesulfonamide (Compound 1121
Figure imgf000048_0003
[0178] Compounds 112 was prepared by directly reduction of rosuvastatin with sodium borohydride. [M+H]+ calculated for C22H30FN3O5S: 468.20; found: 468.2.
Example 10
[0179] N-(5-(( S.E)-4-( ( S V 1.3-Dioxan-4-vlV 3-hvdroxvbut- 1 -en- 1 -vl)-4-(4- fluoroDhenvl)-6-isoDroDvlDvrimidin-2-vl)-N-methvhnethanesulfonamide (Compound 113)
Figure imgf000049_0002
[0180] Compounds 113 was prepared according to a standard acetyl procedure from Compound 112 and formaldehyde. [M+H]+ calculated for C23H30FN3O5S: 480.20; found: 480.2.
Example 11
[0181] N-(4-(4-Fluorophenvl)-5-(fE)-2-((4S.6S)-6-(2-hvdroxvethvl)-l.3-dioxan- 4-vl)vinvll-6-isopropvlpvrimidin-2-vlVN-methvlmethanesulfonamide (Compound 1141
Figure imgf000049_0001
[0182] Compounds 114 was isolated from the reaction in Example 10 to prepare Compound 113 according to a standard acetyl procedure from Compound 112 and formaldehyde. [M+H]~ calculated for C23H30FN3O5S: 480.20; found: 480.2.
Example 12
[0183] 2-((4R-6S)-6-((E)-2-(4-(4-FluoroDhenvl)-6-isopropvl-2-(N- methvlmethvlsulfonamido)pvrimidin-5-vl)vinvl)- 1.3-dioxan-4-vl)acetic acid (Compound
115)
Figure imgf000050_0004
[0184] Compounds 115 was prepared according to Scheme I from rosuvastatin and formaldehyde. [M+H]+ calculated for C23H28FN3O6S: 494.18; foimd: 494.3.
Example 13
[0185] 2-ff4R-6Sl-6 2-f4-M-Fluorophenvn-6-isopropvl-2-iN-
Figure imgf000050_0001
methvlmethvlsulfonamidolt?vrimidin-5-vllvinvlV2-methvl-1.3-dioxan-4-vllacetic acid
(Compound 1161
Figure imgf000050_0005
[0186] Compoimds 116 was prepared according to Scheme I from rosuvastatin and acetaldehyde. [M+H]+ calculated for C24H30FN3O6S: 508.19; found: 508.1.
Example 14
[0187] 2-((4R-6Sl-6-( 2-(4-(4-FluorophenvlV6-isopropvl-2-(N-
Figure imgf000050_0002
methvlmethvlsulfonamidolPYrimidin-5-vlWinvlV2-(pvridin-3- 1.3-dioxan-4-vriacetic acid
Figure imgf000050_0003
(Compound 1171
Figure imgf000050_0006
[0188] Compounds 117 was prepared according to Scheme I from rosuvastatin and nicotinaldehyde. [M+H]+ calculated for C28H31FN4O6S: 571.20; found: 571.1. Example 15
[0189] 2-((4R.6S)-2-(2-AcetoxvphenvD-6-((E)-2-(4-(4-fluorophenvl)-6- isopropvl-2-(N-methvlmethvlsulfonamido)pvrimidin-5-vDvinvD-1.3-dioxan-4-vl)acetic acid (Compound 118)
Figure imgf000051_0001
[0190] Compounds 118 was prepared according to Scheme I from rosuvastatin and nicotinaldehyde. [M+H]+ calculated for C31H34FN3O8S: 628.22; found: 628.1.
Example 16
[0191] 2-((4R.6S)-6-((E)-2-(4-(4-Fluorophenvl)-6-isopropvl-2-(N- methvlmethvlsulfonamido)pvrimidin-5-vl)vinvl)-2-(3-hvdroxvpropvl)-1.3-dioxan-4-vl)acetic acid (Compound 119)
Figure imgf000051_0002
[0192] Compounds 119 was prepared according to Scheme I from rosuvastatin and 4-hydroxybutanal. [M+H]+ calculated for C26H34FN3O7S: 552.22; found: 552.5.
Example 17
[0193] N-(5-((3S.5R.E)-7.7-Diethoxv-3-5-dihvdroxvhept-l-en-l-vl)-4-(4- fluorophenvD-6-isopropvlpvrimidin-2-vD-N-methvlmethanesulfonamide ( Compound 1201
Figure imgf000052_0002
[0194] Compounds 120 was prepared according to Scheme I from rosuvastatin and ethanol in a manner similar to that of Compound 101. [M+H]~ calculated for
C26H38FN3O6S: 540.26; found: 540.2.
Example 18
[0195] (3R.5S.E)-3-(Ethoxvmethoxv)-7-(4-(4-fluorophenvl)-6-isopropvl-2-(N- methvlmethvlsulfonamido)pvrirnidin-5-vl)-5-hvdroxvhept-6-enoic acid (Compound 1211
Figure imgf000052_0003
[0196] Compounds 121 was prepared according to a standard procedure from rosuvastatin and chloromethyl ethyl ether. [M+H]~ calculated for C25H34FN3O7S: 540.22; found: 540.2.
Example 19
[0197] Methvl f 3R.5 S .E)-3 -1 f 4-( 4-fluorophenvl)-6-isoproDvl-
Figure imgf000052_0001
2-fN-methvlmethvlsulfonamido)ovrimidin-5-vl)-5-hvdroxvheot-6-enoate (Compound 122)
Figure imgf000052_0004
[0198] Compounds 122 was prepared according to a standard procedure from rosuvastatin and chloromethyl ethyl ether. [M+H] 1 calculated for C26H36FN3O7S: 554.24; found: 554.2.
Example 20
[0199] f3R.5S.EV5-rEthoxvmethoxvV7-(4-(4-fluoroDhenvlV6-isoDropvl-2-(N- methvlmethvlsulfonamidolpvrimidin-5-vlV3-hvdroxvhept-6-enoic acid (Compound 123)
Figure imgf000053_0001
[0200] Compounds 123 was prepared according to a standard procedure from rosuvastatin and chloromethyl ethyl ether. [M+H] 1 calculated for C25H34FN3O7S: 540.22; found: 540.2.
Example 21
[0201] Methyl f3R.5S.EV5-fethoxvmethoxvl-7-f4-f4-fluorophenvlV6-isopropvl- 2-fN-methvlmethvlsulfonamidolpvrimidin-5-vlV3-hvdroxvhept-6-enoate (Compound 1241
Figure imgf000053_0002
[0202] Compounds 124 was prepared according to a standard procedure from rosuvastatin and chloromethyl ethyl ether. [M+H] 1 calculated for C26H36FN3O7S: 554.24; found: 554.2.
Example 22
[0203] ( 3R.5 S.EV3.5-bisfEthoxvmethoxvV7-(4-(4-fluorophenvlV6- isopropyl- 2- fN-methvlmethvlsulfonamidolpvrimidin-5-vllhept-6-enoic acid ("Compound 1251
Figure imgf000054_0003
[0204] Compounds 125 can be prepared according to a standard procedure from rosuvastatin and chloromethyl ethyl ether. [M+H]+ calculated for CasHioFNgOeS: 598.26.
Example 23
Figure imgf000054_0001
[0206] Compounds 126 was prepared according to a standard procedure from rosuvastatin and chloromethyl ethyl ether. [M+H]~ calculated for C25H32FN3O6S: 522.21; found: 522.2.
Example 24
[0207] N-( 5-((3 S.5S.E)-7-fl3thoxvmethoxv)-3.5-dihvdroxvhept-l -en- 1 -vl)-4-(4- fluoroDhenvl)-6-isoDroDvltrvrimidin-2-vl )-N-methvlmethanesulfonamide f Compound 127)
Figure imgf000054_0002
[0208] Compounds 127 can be prepared according to a standard procedure from rosuvastatin and chloromethyl ethyl ether. [M+H]+ calculated for CisHgeFNsOeS: 526.24.
Example 25
Tissue Distribution Following Oral Administration
[0209] The liver specificity of the disclosed compounds were determined and compared with their corresponding active drug compounds. The disclosed compounds and reference active drug compounds were administered at 5-20 mg/kg to fasted rats by oral gavage. Plasma concentrations of the active, metabolite, and the prodrug compounds in circulation and in the hepatic portal vein were determined by a standard HPLC-UV method. Concentrations of the same in the liver, small intestine, and other organs were measured by the standard LC-MS method.
[0210] Table 1 summarizes the results, which demonstrates improved liver distribution of the disclosed compounds relative to their active drug counterparts. l i l d f /k Ad i i i i
Figure imgf000055_0001

Claims

WHAT IS CLAIMED IS:
1. A compound having the structure of Formula I:
Figure imgf000056_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is selected from the group consisting of optionally substituted Ci-ealkyl, -COOR5, optionally substituted Ca-ioalkoxyalkyl, and optionally substituted 3-12 membered heterocyclyl, or alternatively:
(i) R1 and R2 together with the atoms to which they are attached form an optionally substituted 4-10 membered heterocyclyl, or
(ii) R1 and R3 together with the atoms to which they are attached form an optionally substituted 6-10 membered heterocyclyl;
R2 is selected from the group consisting of H, -C(0)R6, and optionally substituted C2- loalkoxyalkyl, or alternatively:
(i) R1 and R2 together with the atoms to which they are attached form an optionally substituted 4-10 membered heterocyclyl, or
(ii) R2 and R3 together with the atoms to which they are attached form a 6- 10 membered heterocyclyl, optionally substituted with one or more R7; R3 is selected from the group consisting of H, -C(0)R8, and optionally substituted C2- loalkoxyalkyl, or alternatively:
(i) R1 and R3 together with the atoms to which they are attached form an optionally substituted 6-10 membered heterocyclyl, or
(ii) R2 and R3 together with the atoms to which they are attached form a 6- 10 membered heterocyclyl, optionally substituted with one or more R7; R4 is a fragment of a therapeutic agent;
each of R5, R6, and R8 is independently selected from the group consisting of H, optionally substituted Ci-ealkyl, halogen, optionally substituted Ca-ealkenyl, optionally substituted C2-6alkenyl, optionally substituted Ca-vcarbocyclyl, optionally substituted C6- loaryl, optionally substituted 5-10 membered heteroaryl, and optionally substituted 3-10 membered heterocyclyl; and
each R7 is independently selected from the group consisting of optionally substituted Ci-ealkyl, oxo, optionally substituted C6-isaiyl, and optionally substituted 5-18 membered heteroaryl,
wherein at least one of R2 and R3 is not H, or R1 is not -COOH, and
wherein if R1 and R3 together with the atoms to which they are attached form a 6 membered heterocyclyl, then R2 is not H.
2. The compound of claim 1 having the defined 1,3-cis-stereochemistry shown in Formula la or lb:
Figure imgf000057_0001
or a pharmaceutically acceptable salt thereof.
3. The compound of claim 1 having the structure of Formula Ic:
Figure imgf000057_0002
or a pharmaceutically acceptable salt thereof, wherein X is an optionally substituted Ci- valkylene linker.
4. The compound of claim 3 wherein X is a Ci-7alkylene linker optionally substituted with one or more R11, and wherein Rn is selected from the group consisting of Ci-ealkyl, Ci-ealkenyl, Ci-ealkynyl, Ci-eheteroalkyl, Cs-vcarbocyclyl, 3-10 membered heterocyclyl, C6-iearyl, C6-iearylCi-ealkyl, 5-10 membered heteroaryl, 5-10 membered heteroarylCi-ealkyl, halo, cyano, hydroxy, Ci-ealkoxy, Ca-ioalkoxyalkyl, C6-iearyloxy, sulfhydryl, haloCi-ealkyl, haloCi-ealkoxy, Ci-ealkylthio, C6-iearylthio, and nitro.
5. The compound of claim 3 or 4, wherein R11 is selected from the group consisting of Ci-6alkyl, C3-7carbocyclyl, Ci-ealkoxy, C6-isaryl, and halo.
6. The compound of any one of claims 3-5, wherein R11 is selected from the group consisting of Ci-ealkyl and C6-isaryl.
7. The compound of any one of claims 3-6, wherein X is a Caalkylene linker.
8. The compound of any one of claims 3-7, wherein X is -CH2CH2-.
9. The compound of any one of claims 3-6, wherein X is a Csalkylene linker.
10. The compound of any one of claims 3-6 or 9, wherein X is -CH2CH2CH2-.
11. The compound of any one of claims 3-6 or 9, wherein X is -CH2(CH2CH3)CH2-.
12. The compound of any one of claims 3-6 or 9, wherein X is -
Figure imgf000058_0002
13. The compound of claim 1 having the structure of Formula Id:
Figure imgf000058_0001
or a pharmaceutically acceptable salt thereof, wherein each of R9 and R10 is independently an optionally substituted Ci-ealkyl.
14. The compound of claim 13 wherein each of R9 and R10 is independently a Ci- ealkyl optionally substituted with one or more R12, and wherein R12 is selected from the group consisting of Ci-ealkyl, Ci-ealkenyl, Ci-ealkynyl, Ci-eheteroalkyl, Cs-vcarbocyclyl, C3- 7carbocyclylCi-6alkyl, 3-10 membered heterocyclyl, 3-10 membered heterocyclylCi-ealkyl, C6-iearyl, C6-isarylCi-ealkyl, 5-10 membered heteroaryl, 5-10 membered heteroaryl C 1 -ealkyl, and haloCi-ealkyl.
15. The compound of claim 13 or 14 wherein R12 is selected from the group consisting of Ci-ealkyl, Ci-eheteroalkyl, C6-iearyl, C6-isarylCi-ealkyl, and haloCi-ealkyl.
16. The compound of any one of claims 13-15, wherein R9 is a Ci-ealkyl.
17. The compound of any one of claims 13-16, wherein R9 is ethyl.
18. The compound of any one of claims 13-17, wherein R10 is a Ci-ealkyl.
19. The compound of any one of claims 13-18, wherein R10 is ethyl.
20. The compound of claim 1 having the structure of Formula Ie:
Figure imgf000059_0003
or a pharmaceutically acceptable salt thereof.
21. The compound of claim 1 having the structure of Formula If:
Figure imgf000059_0001
or a pharmaceutically acceptable salt thereof, wherein Y is a Ci-salkylene linker, optionally substituted with one or more R7.
22. The compound of claim 21 wherein R7 is selected from the group consisting of optionally substituted C6-isaryl and optionally substituted 5-18 member ed heteroaryl.
23. The compound of claim 21 or 22 wherein Y is a Cialkylene linker.
24. The compound of claim 1 having the structure of Formula Ig:
Figure imgf000059_0002
or a pharmaceutically acceptable salt thereof.
25. The compound of any one of claims 1-2 or 21-23, wherein R1 is an optionally substituted Ci-salkyl.
26. The compound of any one of claims 1-2, 21-23, or 25, wherein R1 is a Ci-salkyl optionally substituted with one or more R13, and wherein R13 is selected from the group consisting of halo, cyano, hydroxy, Ci-ealkoxy, C6-ioaryloxy, sulfhydryl, haloCi-ealkoxy, Ci- ealkylthio, C6-ioarylthio, and nitro.
27. The compound of any one of claims 1-2, 21-23, or 25-26, wherein R13 is selected from the group consisting of halo, hydroxy, Ci-ealkoxy, and C6-ioaryloxy.
28. The compound of any one of claims 1-2, 21-23, or 25-27, wherein R1 is -CH2OH.
29. The compound of any one of claims 1-2 or 21-23, wherein R1 is -COOR5.
30. The compound of any one of claims 1-2, 21-23, or 29, wherein R5 is H.
31. The compound of any one of claims 1-2, 21-23, or 29, wherein R5 is methyl.
32. The compound of any one of claims 1-2 or 21-23, wherein R1 is an optionally substituted Ca-ioalkoxy alkyl.
33. The compound of any one of claims 1-2, 21-23, or 32, wherein R1 is a C2- loalkoxyalkyl optionally substituted with one or more R14, and wherein R14 is selected from the group consisting of halo, cyano, hydroxy, Ci-ealkoxy, C6-ioaryloxy, sulfhydryl, haloCi- ealkoxy, C1-6alkylthio, C6-10arylthio, and nitro.
34. The compound of any one of claims 1-2, 21-23, or 32-33, wherein R14 is selected from the group consisting of halo, hydroxy, Ci-ealkoxy, and C6-ioaryloxy.
35. The compound of any one of claims 1-2, 21-23, or 32-34, wherein R1 is - CH2OCH2OCH2CH3.
36. The compound of any one of claims 1-2, 21-23, or 32-34, wherein R1 is - CH(OCH2CH3)2.
37. The compound of any one of claims 1-2 or 21-23, wherein R1 is an optionally substituted 3-12 member ed heterocyclyl.
38. The compound of any one of claims 1-2, 21-23, or 37, wherein R1 is a 3-12 membered heterocyclyl optionally substituted with one or more R15, and wherein R15 is selected from the group consisting of Ci-ealkyl, Ci-ealkenyl, Ci-ealkynyl, Ci-eheteroalkyl, C3-7carbocyclyl, 3-10 membered heterocyclyl, C6-isaryl, C6-isarylCi-ealkyl, 5-10 membered heteroaryl, 5-10 membered heteroarylCi-ealkyl, halo, cyano, hydroxy, Ci-6alkoxy, C2- loalkoxyalkyl, aryloxy, sulfhydryl, haloCi-ealkyl, haloCi-ealkoxy, Ci-ealkylthio, C6- lsarylthio, and nitro.
39. The compound of any one of claims 1-2, 21-23, or 37-38, wherein R15 is selected from the group consisting of Ci-ealkyl, C3-7carbocyclyl, Ci-ealkoxy, C6-isaryl, and halo.
40. The compound of any one of claims 1-2, 21-23, or 37-39, wherein R1 is a 5 membered heterocyclyl.
41. The compound of any one of claims 1-2, 21-23, or 37-40, wherein R1 is
Figure imgf000061_0001
42. The compound of any one of claims 1-2, 21-23, or 37-39, wherein R1 is a 6 membered heterocyclyl.
43. The compound of any one of claims 1-2, 21-23, 37-39, or 42, wherein R1 is
Figure imgf000061_0002
44. The compound of any one of claims 1-20, wherein R2 is H.
45. The compound of any one of claims 1-20 or 24, wherein R2 is -C(0)R6.
46. The compound of any one of claims 1-20, 24, or 45, wherein R6 is an optionally substituted Ci^alkyl.
47. The compound of any one of claims 1-20, 24, or 45-46, wherein R6 is a Ci-ealkyl optionally substituted with one or more R16, and wherein R16 is selected from the group consisting of halo, cyano, hydroxy, Ci-ealkoxy, C6-ioaryl, C6-ioaryloxy, sulfhydryl, haloCi- ealkoxy, Ci-ealkylthio, C6-ioarylthio, and nitro.
48. The compound of any one of claims 1-20, 24, or 45-47, wherein R16 is selected from the group consisting of halo, hydroxy, Ci-ealkoxy, C6-ioaryl, and C6-ioaryloxy.
49. The compound of any one of claims 1-20, 24, or 45-48, wherein R6 is -CH(CH3)2.
50. The compound of any one of claims 1-20 or 24, wherein R2 is an optionally substituted C2-ioalkoxyalkyl.
51. The compound of any one of claims 1-20, 24, or 50, wherein R2 is a C2- loalkoxyalkyl optionally substituted with one or more R17, and wherein R17 is selected from the group consisting of halo, cyano, hydroxy, Ci-ealkoxy, C6-ioaryl, C6-ioaryloxy, sulfhydryl, haloCi-ealkoxy, Ci-ealkylthio, C6-ioarylthio, and nitro.
52. The compound of any one of claims 1-20, 24, or 50-51, wherein R17 is selected from the group consisting of halo, hydroxy, Ci-ealkoxy, C6-ioaryl, and C6-ioaryloxy.
53. The compound of any one of claims 1-20, 24, or 50-52, wherein R2 is - CH2OCH2CH3.
54. The compound of any one of claims 1-20, wherein R3 is H.
55. The compound of any one of claims 1-20, wherein R3 is -C(0)R8.
56. The compound of any one of claims 1-20 or 55, wherein R8 is an optionally substituted Ci-ealkyl.
57. The compound of any one of claims 1-20 or 55-56, wherein R8 is a Ci-ealkyl optionally substituted with one or more R18, and wherein R18 is selected from the group consisting of halo, cyano, hydroxy, Ci-ealkoxy, C6-ioaryl, C6-ioaryloxy, sulfhydryl, haloCi- ealkoxy, Ci-ealkylthio, C6-ioarylthio, and nitro.
58. The compound of any one of claims 1-20 or 55-57, wherein R18 is selected from the group consisting of halo, hydroxy, Ci-ealkoxy, C6-ioaryl, and C6-ioaryloxy.
59. The compound of any one of claims 1-20 or 55-58, wherein R8 is -CH(CH3)2.
60. The compound of any one of claims 1-20, wherein R3 is an optionally substituted C2-10alkoxyalkyl.
61. The compound of any one of claims 1-20 or 60, wherein R3 is a C2-ioalkoxyalkyl optionally substituted with one or more R19, and wherein R19 is selected from the group consisting of halo, cyano, hydroxy, Ci-ealkoxy, C6-ioaryl, C6-ioaryloxy, sulfhydryl, haloCi- 6alkoxy, C1-6alkylthio, C6-ioarylthio, and nitro.
62. The compound of any one of claims 1-20 or 60-61, wherein R19 is selected from the group consisting of halo, hydroxy, Ci-ealkoxy, C6-ioaryl, and C6-ioaryloxy.
63. The compound of any one of claims 1-20 or 60-62, wherein R3 is -CH2OCH2CH3.
64. The compound of any one of claims 1-23 or 25-43, wherein R7 is an optionally substituted C1-8alkyl.
65. The compound of any one of claims 1-23, 25-43, or 64, wherein R7 is a Ci-salkyl optionally substituted with one or more R20, and wherein R20 is selected from the group consisting of halo, cyano, hydroxy, Ci-ealkoxy, C6-ioaryl, C6-ioaryloxy, sulfhydryl, haloCi- ealkoxy, C1-6alkylthio, C6-ioarylthio, and nitro.
66. The compound of any one of claims 1-23, 25-43, or 64-65, wherein R20 is selected from the group consisting of halo, hydroxy, Ci-ealkoxy, C6-ioaryl, and C6-ioaryloxy.
67. The compound of any one of claims 1-23, 25-43, or 64-66, wherein R7 is methyl.
68. The compound of any one of claims 1-23, 25-43, or 64-66, wherein R7 is - CH2CH2CH2OH.
69. The compound of any one of claims 1-23 or 25-43, wherein R7 is oxo.
70. The compound of any one of claims 1-23 or 25-43, wherein R7 is an optionally substituted C6-iearyl.
71. The compound of any one of claims 1-23, 25-43, or 70, wherein R7 is a C6-18aryl optionally substituted with one or more R21.
72. The compound of any one of claims 1-23, 25-43, or 70-71, wherein R21 is selected from the group consisting of Ci-ealkyl, halo, cyano, hydroxy, Ci-6alkoxy, haloCi-ealkyl, haloCi-ealkoxy, nitro, -C(=0)NR22R23, -N(R24)C(=O)R25, -C(=0)0R26, -0C(=0)R27, and - C(=0)R28, and wherein each of R22, R23, R24, R25, R26, R27, and R28 is independently selected from the group consisting of hydrogen, Ci-ealkyl, C2-6alkenyl, C2-6alkenyl, C3-7carbocyclyl, Gs-ioaryl, 5-10 membered heteroaryl, and 3-10 membered heterocyclyl.
73. The compound of any one of claims 1-23, 25-43, or 70-72, wherein each of R22, R23, R24, R25, R26, R27, and R28 is independently selected from the group consisting of hydrogen, Ci-ealkyl, C2-6alkenyl, and C6-ioaryl.
74. The compound of any one of claims 1-23, 25-43, or 70-73, wherein R7 is a phenyl.
75. The compound of any one of claims 1-23, 25-43, or 70-74, wherein R7 is
Figure imgf000063_0001
76. The compound of any one of claims 1-23 or 25-43, wherein R7 is an optionally substituted 5-18 member ed heteroaryl.
77. The compound of any one of claims 1-23, 25-43, or 76, wherein R7 is a 5-18 membered heteroaryl optionally substituted with one or more R29.
78. The compound of any one of claims 1-23, 25-43, or 76-77, wherein R29is selected from the group consisting of Ci-ealkyl, halo, cyano, hydroxy, Ci-ealkoxy, haloCi-ealkyl, haloCi-ealkoxy, nitro, -C(=O)NR30R31, -N(R32)C(=0)R33, -C(=0)0R34, -0C(=O)R35, and - C(=0)R36, and wherein each of R30, R31, R32, R33, R34, R35, and R36 is independently selected from the group consisting of hydrogen, Ci-ealkyl, C2-6alkenyl, Ci-ealkynyl, C3-7carbocyclyl, C6-ioaryl, 5-10 membered heteroaiyl, and 3-10 membered heterocyclyl.
79. The compound of any one of claims 1-23, 25-43, or 76-78, wherein each of R30, R31, R32, R33, R34, R35, and R36 is independently selected from the group consisting of hydrogen, Ci-ealkyl, C2-6alkenyl, and C6-ioaryl.
80. The compound of any one of claims 1-23, 25-43, or 76-79, wherein R7 is a 6 membered heteroaryl.
81. The compound of any one of claims 1-23, 25-43, or 76-80, wherein R7 is
Figure imgf000064_0001
82. The compound of any one of claims 1-81, wherein R4 is a fragment of a livertargeting therapeutic agent or a fragment of a liver-activated therapeutic agent.
83. The compound of any one of claims 1-82, wherein R4 is a fragment of a lipidlowering therapeutic agent or a fragment of a cholesterol-biosynthesis-inhibiting therapeutic agent
84. The compound of any one of claims 1-83, wherein R4 is a fragment of a HMG- CoA reductase inhibitor.
85. The compound of any one of claims 1-84, wherein R4 is a fragment of a statin.
86. The compound of any one of claims 1-85, wherein R4 is selected from the group
consisting of
Figure imgf000064_0002
Figure imgf000065_0001
87. The compound of any one of claims 1-86, wherein R4 is
Figure imgf000066_0001
88. A compound having the structure of Formula P:
Figure imgf000066_0002
or a pharmaceutically acceptable salt thereof, wherein:
Z is O, S, or NR40;
R37 is selected from the group consisting of H, a fragment of a therapeutic agent, optionally substituted Ci-ealkyl, Ci-ehaloalkyl, Ci-eheteroalkyl, optionally substituted C3- ?carbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted G>- learyl, 5-10 membered heteroaryl, and optionally substituted C2-ioalkoxyalkyl;
each of R38 and R39 is independently selected from the group consisting of a fragment of a therapeutic agent, optionally substituted Ci-ealkyl, Ci-ehaloalkyl, Ci-eheteroalkyl, optionally substituted C3-7carbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C6-iearyl, optionally substituted Ca-ioalkoxyalkyl, and optionally substituted 5-10 membered heteroaryl, wherein at least one of R37, R38, and R39 is a fragment of a therapeutic agent, wherein the fragment of a therapeutic agent is further optionally substituted with one
or more moieties selected from the group consisting of
Figure imgf000067_0002
Figure imgf000067_0001
i-ealkyl, halo, and C6-18aryl;
each A is independently selected from O, S, and NR43, provided that at least one A is
O;
R40 is independently selected from the group consisting of optionally substituted Ci- ealkyl, Ci-ehaloalkyl, C1-6heteroalkyl, optionally substituted C3-?carbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C6-isaryl, optionally substituted C2-10alkoxyalkyl, and optionally substituted 5-10 membered heteroaryl; and
each of R41, R42, and R43 is independently selected from the group consisting of H, optionally substituted C1-6alkyl, Ci-ehaloalkyl, Ci-eheteroalkyl, optionally substituted C3- vcarbocyclyl, optionally substituted 3-10 membered heterocyclyl, optionally substituted C6- lgaryl, optionally substituted C2-10alkoxyalkyl, and optionally substituted 5-10 membered heteroaryl.
89. The compound claim 88, wherein Z is O.
90. The compound of claim 88 or 89, wherein R37 is H.
91. The compound of any one of claims 88-90, wherein R38 is the optionally substituted fragment of a therapeutic agent.
92. The compound of any one of claims 88-91, wherein the fragment of a therapeutic
agent is
Figure imgf000067_0003
93. The compound of any one of claims 88-92, wherein R39 is ethyl.
94. The compound of any one of claims 91-93, wherein the fragment of a therapeutic agent is substituted with methyl, such that R38 has the structure
Figure imgf000068_0002
95. The compound of any one of claims 88-91, wherein the fragment of a therapeutic agent is:
Figure imgf000068_0001
96. The compound of claim 95, wherein each A is O.
97. The compound of claim 95 or 96, wherein R41 is H.
98. The compound of any one of claims 95-97, wherein R42 is ethyl.
99. The compound of claim 1, selected from the group consisting of
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
100. A pharmaceutical composition comprising a therapeutically effective amount of a compound of any one of claims 1-99, and a pharmaceutically acceptable excipient
101. The pharmaceutical composition of claim 100, further comprising an additional medicament.
102. The pharmaceutical composition of claim 101, wherein the additional medicament is selected from the group consisting of a cholesterol absorption inhibitor, a hypertension medicament niacin, and aspirin.
103. The pharmaceutical composition of claim 101 or 102, wherein the cholesterol absorption inhibitor is ezetimibe.
104. The pharmaceutical composition of claim 101 or 102, wherein the hypertension medicament is selected from the group consisting of a diuretic, a beta-blocker, an angiotensin-converting enzyme inhibitor, an angiotensin receptor blocker, a calcium channel blocker, an alpha blocker, an alpha-2 receptor agonist a combined alpha and beta- blocker, a central agonist a peripheral adrenergic inhibitor, and a vasodilator.
105. The pharmaceutical composition of claim 101, 102, or 104, wherein the hypertension medicament is selected from the group consisting of acebutolol, alpha methyldopa, amiloride hydrochloride, amlodipine besylate, atenolol, benazepril hydrochloride, betaxolol, bepridil, bisoprolol fumarate, bumetanide, candesartan, captopril, carteolol hydrochloride, carvedilol, chlorthalidone, chlorothiazide, clonidine hydrochloride, diltiazem hydrochloride, doxazosin mesylate, enalapril maleate, eprosartan mesylate, felodipine, fosinopril sodium, furosemide, hydralazine hydrochloride, hydrochlorothiazide, guanabenz acetate, guanadrel, guanethidine monosulfate, guanfacine hydrochloride, indapamide, irbesarten, isradipine, labetalol hydrochloride, lisinopril, losartan potassium, methyldopa, metolazone, metoprolol succinate, metoprolol tartrate, minoxidil, moexipril, nadolol, nicardipine, nifedipine, nisoldipine, penbutolol sulfate, perindopril, pindolol, prazosin hydrochloride, propranolol hydrochloride, quinapril hydrochloride, ramipril, reserpine, telmisartan, terazosin hydrochloride, trandolapril, triamterene, solotol hydrochloride, spironolactone, timolol maleate, valsartan, and verapamil hydrochloride.
106. A method of treating a liver disease, disorder, or condition, comprising administering a compound of any one of claims 1-99 or administering a pharmaceutical composition of any one of claims 100-105, to a subject in need thereof.
107. The method of claim 106, wherein the liver disease, disorder, or condition involves the metabolism, storage, or homeostatic control of cholesterol, fatty acids, bile, triglycerides, plasma proteins, carrier proteins, lipoproteins, acute phase proteins, apolipoproteins, carbohydrates, hemostatic factors, fibrinolysis factors, thrombopoietin, insulin-like growth factor 1, hepcidin, angiotensinogen, vitamin A, vitamin D, vitamin B12, vitamin K, iron, copper, or catalase.
108. The method of claim 106 or 107, wherein the liver disease, disorder, or condition is selected from the group consisting of liver fibrosis, cirrhosis, hepatitis, alcoholic liver disease, fatty liver, liver cancer, hemochromatosis, Wilson’s disease, primary sclerosing cholangitis, alpha 1 -antitrypsin deficiency, hyperglycemia, hypoglycemia, and diabetes.
109. A method of treating a cardiovascular disease, disorder, or condition, comprising administering a compound of any one of claims 1-99 or administering a pharmaceutical composition of any one of claims 100-105, to a subject in need thereof.
110. The method of claim 109, wherein the cardiovascular disease, disorder, or condition is dyslipidemia or atherosclerosis.
111. The method of claim 109 or 110, wherein the cardiovascular disease, disorder, or condition is hyperlipidemia.
112. A method of altering blood lipid concentration, comprising administering a compound of any one of claims 1-99 or administering a pharmaceutical composition of any one of claims 100-105, to a subject in need thereof.
113. A method of inhibiting HMG-CoA reductase, comprising administering a compound of any one of claims 1-99 or administering a pharmaceutical composition of any one of claims 100-105, to a subject in need thereof.
114. The method of any one of claims 106-114, wherein the subject is a mammal.
115. The method of any one of claims 106-114, wherein the subject is a human.
PCT/US2019/012762 2018-01-09 2019-01-08 Acetal compounds and therapeutic uses thereof WO2019139919A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
KR1020207022964A KR20200123106A (en) 2018-01-09 2019-01-08 Acetal compounds and therapeutic uses thereof
CN201980016007.3A CN111788196A (en) 2018-01-09 2019-01-08 Acetal compounds and their therapeutic use
RU2020126177A RU2020126177A (en) 2018-01-09 2019-01-08 ACETAL COMPOUNDS AND THEIR THERAPEUTIC APPLICATIONS
US16/960,681 US11970482B2 (en) 2018-01-09 2019-01-08 Acetal compounds and therapeutic uses thereof
CA3087932A CA3087932A1 (en) 2018-01-09 2019-01-08 Acetal compounds and therapeutic uses thereof
EP19738393.8A EP3737676B1 (en) 2018-01-09 2019-01-08 Acetal compounds and therapeutic uses thereof
JP2020537621A JP2021509907A (en) 2018-01-09 2019-01-08 Acetal compounds and their therapeutic use
AU2019207625A AU2019207625A1 (en) 2018-01-09 2019-01-08 Acetal compounds and therapeutic uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862615357P 2018-01-09 2018-01-09
US62/615,357 2018-01-09

Publications (1)

Publication Number Publication Date
WO2019139919A1 true WO2019139919A1 (en) 2019-07-18

Family

ID=67218355

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/012762 WO2019139919A1 (en) 2018-01-09 2019-01-08 Acetal compounds and therapeutic uses thereof

Country Status (9)

Country Link
US (1) US11970482B2 (en)
EP (1) EP3737676B1 (en)
JP (1) JP2021509907A (en)
KR (1) KR20200123106A (en)
CN (1) CN111788196A (en)
AU (1) AU2019207625A1 (en)
CA (1) CA3087932A1 (en)
RU (1) RU2020126177A (en)
WO (1) WO2019139919A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4005633A2 (en) 2022-02-08 2022-06-01 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester
WO2023152754A1 (en) * 2022-02-08 2023-08-17 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106061984A (en) 2014-02-13 2016-10-26 配体药物公司 Prodrug compounds and their uses
CN113292542A (en) * 2021-05-26 2021-08-24 乳源东阳光药业有限公司 Rosuvastatin calcium intermediate impurity and preparation method thereof

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987005297A1 (en) 1986-03-03 1987-09-11 The University Of Chicago Cephalosporin derivatives
WO2007125547A2 (en) * 2006-05-03 2007-11-08 Manne Satyanarayana Reddy Novel process for statins and its pharmaceutically acceptable salts thereof
KR20100087931A (en) * 2009-01-29 2010-08-06 (주)유케이케미팜 Precursive compound of rosuvastatin and manufacturing method for the precursive compound of rosuvastatin
WO2010103320A1 (en) * 2009-03-10 2010-09-16 Bradford Pharma Limited Use of rosuvastatin lactols as medicaments
CN102079726A (en) * 2009-11-27 2011-06-01 上海医药工业研究院 Miazine compounds, intermediates of miazine compounds, preparation method of intermediates and miazine compounds as well as application of miazine compound
WO2012038785A1 (en) * 2010-09-21 2012-03-29 Biocon Limited Polymorphs of rosuvastatin acetonide calcium ((3r,5s,6e)-7-[4-(4- fluorophenyl)-6-isopropyl-2-(methanesulfonyl-methyl-amino)-pyrimn)in-5- yl)vinyl)-2,2-dimethyl-l,3-dioxan-4-yl) acetic acid calcium salt
US8207333B2 (en) * 2005-06-24 2012-06-26 Lek Pharmaceuticals D. D. Process for preparing pure amorphous rosuvastatin calcuim
US8354530B2 (en) * 2005-07-28 2013-01-15 Lek Pharmaceuticals d. d Process for the synthesis of rosuvastatin calcium
KR20160126700A (en) * 2015-04-24 2016-11-02 미래파인켐 주식회사 New Statin intermediate, the preparation of the same and the preparation of Rosuvastatin using the same
US20170183314A1 (en) * 2013-11-25 2017-06-29 Fudan University Method for preparing rosuvastatin sodium
KR20170078033A (en) * 2015-12-29 2017-07-07 미래파인켐 주식회사 Novel Statin intermediate, the preparation method of the same and the preparation method of Rosuvastatin using the same
WO2017137469A1 (en) * 2016-02-11 2017-08-17 Stichting Katholieke Universiteit Novel class of compounds for the treatment of cardiovascular disease
CN107382875A (en) * 2017-06-26 2017-11-24 浙江美诺华药物化学有限公司 A kind of synthetic method of rosuvastain calcium chiral isomer impurity

Family Cites Families (279)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE563332A (en) 1956-12-20
US3116282A (en) 1960-04-27 1963-12-31 Upjohn Co Pyrimidine nucleosides and process
CA777769A (en) 1963-03-18 1968-02-06 H. Roy Clarence Substituted methylene diphosphonic acid compounds and detergent compositions
US3328388A (en) 1964-09-02 1967-06-27 Merck & Co Inc Arabinofuranosyl pyrimidines and methods of preparing same
CH492738A (en) 1967-09-13 1970-06-30 Sandoz Ag Chlorethyl group-containing oxazaphosph- - orine derivatives
BE768925A (en) 1970-06-30 1971-11-03 Takeda Chemical Industries Ltd ADENOSINE DERIVATIVES AND METHOD OF PREPARATION
DE2752125A1 (en) 1977-11-22 1979-05-23 Max Planck Gesellschaft NEW ORGANIC PHOSPHORUS RING COMPOUNDS, THEIR PRODUCTION AND USE
US4376165A (en) 1978-06-22 1983-03-08 Miles Laboratories, Inc. Method of preparing an enzyme-labeled ligand for use in specific binding assays and the labeled conjugate produced thereby
US4318982A (en) 1979-06-04 1982-03-09 Miles Laboratories, Inc. FMN-Labeled specific binding assay
US4340668A (en) 1979-06-04 1982-07-20 Miles Laboratories, Inc. Heme-labeled specific binding assay
US4255566A (en) 1980-02-19 1981-03-10 Miles Laboratories Flavin adenine dinucleotide derivatives and labeled conjugates prepared therefrom
JPH0692381B2 (en) * 1980-03-31 1994-11-16 三共株式会社 MB-530A derivative
US4447529A (en) 1981-07-06 1984-05-08 Miles Laboratories, Inc. Preparing homogeneous specific binding assay element to avoid premature reaction
DE3132221A1 (en) 1981-08-14 1983-05-19 Behringwerke Ag, 3550 Marburg NEW CYCLOPHOSPHAMIDE DERIVATIVES, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE
DE3133077A1 (en) 1981-08-21 1983-03-10 Henkel KGaA, 4000 Düsseldorf NEW 1,3,2-OXAZAPHOSPHORINE COMPOUNDS CONTAINING NEW CYTOSTATICALLY EFFECTIVE EPOXY GROUPS, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE IN PHARMACEUTICAL PREPARATIONS
IT1201087B (en) 1982-04-15 1989-01-27 Gentili Ist Spa PHARMACOLOGICALLY ACTIVE BIPPHOSPHONES, PROCEDURE FOR THEIR PREPARATION AND RELATED PHARMACEUTICAL COMPOSITIONS
US4440740A (en) 1982-04-26 1984-04-03 Merck & Co., Inc. α-Keto aldehydes as enhancing agents of gastro-intestinal drug absorption
US4464389A (en) 1982-10-20 1984-08-07 Sri International Esters of retinoic acid and pentaerythritol and monobenzal acetals thereof
CS233665B1 (en) 1983-01-06 1985-03-14 Antonin Holy Processing of isomere o-phosphonylmethylderivative of anantiomere racemic vicinal diene
US5047533A (en) 1983-05-24 1991-09-10 Sri International Acyclic purine phosphonate nucleotide analogs
EP0158057B1 (en) 1984-03-01 1989-04-26 ASTA Pharma AG Salts of oxazaphosphorine derivatives
US4579849A (en) 1984-04-06 1986-04-01 Merck & Co., Inc. N-alkylguanine acyclonucleosides as antiviral agents
FR2562543B1 (en) 1984-04-10 1987-09-25 Elf Aquitaine NOVEL CYCLIC PHOSPHONITES, THEIR PREPARATION AND APPLICATIONS
US4749694A (en) 1984-04-26 1988-06-07 Merck & Co., Inc. Novel lysine esters used as absorption
US4537772A (en) 1984-05-02 1985-08-27 Merck & Co., Inc. Enhancing absorption of drugs from gastrointestinal tract using acylcarnitines
US4898724A (en) 1984-06-04 1990-02-06 The Dow Chemical Company Organis amine phosphonic acid complexes for the treatment of calcific tumors
NL8403224A (en) 1984-10-24 1986-05-16 Oce Andeno Bv DIOXAPHOSPHORINANS, THEIR PREPARATION AND THE USE FOR SPLITTING OF OPTICALLY ACTIVE COMPOUNDS.
IT1196315B (en) 1984-10-29 1988-11-16 Gentili Ist Spa PROCEDURE FOR THE PREPARATION OF DIPHOSPHONIC ACIDS
IL77243A (en) 1984-12-21 1996-11-14 Procter & Gamble Pharmaceutical compositions containing geminal diphosphonic acid compounds and certain such novel compounds
US4724232A (en) 1985-03-16 1988-02-09 Burroughs Wellcome Co. Treatment of human viral infections
CS263951B1 (en) 1985-04-25 1989-05-12 Antonin Holy 9-(phosponylmethoxyalkyl)adenines and method of their preparation
CS263952B1 (en) 1985-04-25 1989-05-12 Holy Antonin Remedy with antiviral effect
US4729989A (en) 1985-06-28 1988-03-08 Merck & Co., Inc. Enhancement of absorption of drugs from gastrointestinal tract using choline ester salts
US4973579A (en) 1985-06-28 1990-11-27 Merck & Co., Inc. Enhancment of absorption of drugs from gastrointestinal tract using choline ester salts
US4822773A (en) 1985-06-28 1989-04-18 Merck & Co., Inc. Enhancement of absorption of drugs from gastrointestinal tract using choline ester salts
US4692441A (en) 1985-08-16 1987-09-08 Merck & Co., Inc. Chorine esters as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal cavities
US4731360A (en) 1985-08-16 1988-03-15 Merck & Co., Inc. Acylcarnitines as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal compartments
US4847298A (en) 1985-08-16 1989-07-11 Merck & Co., Inc. Acylcarnitines as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal compartments
US4963525A (en) 1985-08-16 1990-10-16 Merck & Co., Inc. Acylcarnitines as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal compartments
US4963556A (en) 1985-08-16 1990-10-16 Merck & Co., Inc. Choline esters as absorption-enhancing agents for drug delivery through mucous membranes of the nasal, buccal, sublingual and vaginal cavities
US4757141A (en) 1985-08-26 1988-07-12 Applied Biosystems, Incorporated Amino-derivatized phosphite and phosphate linking agents, phosphoramidite precursors, and useful conjugates thereof
US4879277A (en) 1985-08-26 1989-11-07 The United States Of America As Represented By The Department Of Health And Human Services Antiviral compositions and methods
US5258538A (en) 1985-08-26 1993-11-02 Applied Biosystems, Inc. 2,3-disubstituted-1,3,2-oxazaphosphacycloalkanes as nucleic acid linking agents
JPS62501712A (en) 1985-08-26 1987-07-09 アメリカ合衆国 Anti-HTLV-3/LAV agent containing 2',3'-dideoxyinosine, 2',3'-dideoxyguanosine or 2',3'-dideoxyadenosine
US5246937A (en) 1985-09-18 1993-09-21 Beecham Group P.L.C. Purine derivatives
DE3600289A1 (en) 1986-01-08 1987-07-09 Bayer Ag CYCLIC MALONYLPHOSPHONIC ACID DIAMOND, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE AS A PEST CONTROL
JPH0615553B2 (en) 1986-02-20 1994-03-02 日産化学工業株式会社 Dihydropyridine-5-phosphonamidic acids
US4968788A (en) 1986-04-04 1990-11-06 Board Of Regents, The University Of Texas System Biologically reversible phosphate and phosphonate protective gruops
US4839466A (en) 1986-04-11 1989-06-13 The Rockefeller University Insulin activity messengers
JPS62249996A (en) 1986-04-23 1987-10-30 Asahi Chem Ind Co Ltd Novel benzoxazaphosphorine derivative and insecticidal composition containing said derivative as active ingredient
US5091552A (en) 1986-06-30 1992-02-25 Board Of Regents, The University Of Texas System Novel antitumor aldophosphamide analogs
ES2038623T3 (en) 1986-07-11 1993-08-01 Asta Medica Aktiengesellschaft PROCEDURE TO PREPARE OXAZAPHOSPHORIN SOLUTIONS WITH IMPROVED STABILITY.
CS264222B1 (en) 1986-07-18 1989-06-13 Holy Antonin N-phosphonylmethoxyalkylderivatives of bases of pytimidine and purine and method of use them
DE3626058A1 (en) 1986-08-01 1988-02-11 Boehringer Mannheim Gmbh NEW DIPHOSPHONIC ACID DERIVATIVES, METHOD FOR THE PRODUCTION THEREOF AND MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS
JPS6360929A (en) 1986-08-29 1988-03-17 Yodogawa Seiyaku Kk Antitumor agent
IL84497A (en) 1986-11-21 1994-10-21 Ciba Geigy Ag 2-(Imidazol-1-yl) ethane-1,1-diphosphonic acid derivatives, their preparation and pharmaceutical compositions containing them
DE3850263D1 (en) 1987-01-19 1994-07-21 Ciba Geigy Substituted dioxane derivatives.
US5159067A (en) 1987-01-28 1992-10-27 University Of Georgia Research Foundation Inc. 5'-Diphosphohexose nucleoside pharmaceutical compositions
US5077280A (en) 1988-04-12 1991-12-31 Brown University Research Foundation Treatment of viral infections
EP0338372A3 (en) 1988-04-22 1991-10-09 American Cyanamid Company Solubilized pro-drugs
GB8815265D0 (en) 1988-06-27 1988-08-03 Wellcome Found Therapeutic nucleosides
ATE128623T1 (en) 1988-08-02 1995-10-15 Nissan Chemical Ind Ltd AGENT FOR IMPROVING DRUG EFFECTS FOR ANTITUMOR DRUGS.
US4882142A (en) 1988-12-19 1989-11-21 The Dow Chemical Company Bone marrow suppressing agents
US5658889A (en) 1989-01-24 1997-08-19 Gensia Pharmaceuticals, Inc. Method and compounds for aica riboside delivery and for lowering blood glucose
US5118672A (en) 1989-07-10 1992-06-02 University Of Georgia Research Foundation 5'-diphosphohexose nucleoside pharmaceutical compositions
JPH0377894A (en) 1989-08-18 1991-04-03 Toray Ind Inc Methylene diphosphonic acid compound
US5204466A (en) 1990-02-01 1993-04-20 Emory University Method and compositions for the synthesis of bch-189 and related compounds
US6642245B1 (en) 1990-02-01 2003-11-04 Emory University Antiviral activity and resolution of 2-hydroxymethyl-5-(5-fluorocytosin-1-yl)-1,3-oxathiolane
US5627165A (en) 1990-06-13 1997-05-06 Drug Innovation & Design, Inc. Phosphorous prodrugs and therapeutic delivery systems using same
CS276072B6 (en) 1990-08-06 1992-03-18 Ustav Organicke Chemie A Bioch (2R)-2-/DI(2-PROPYL)PHOSPHONYLMETHOXY/-3-p-TOLUENESULFONYLOXY -1- TRIMETHYLACETOXYPROPANE AND PROCESS FOR PREPARING THEREOF
DE10399025I2 (en) 1990-09-14 2007-11-08 Acad Of Science Czech Republic Active substance precursors of phosphonates
HU206883B (en) 1990-12-18 1993-01-28 Richter Gedeon Vegyeszet Process for producing new 15-nitro-2-beta-, 3-beta-dihydro- and 15-nitro-2-beta, 3-beta-, 6,7-tetrahydro-tabersonin derivatives and pharmaceutical compositions containing them
US5168103A (en) 1991-01-22 1992-12-01 American Home Products Corporation [[2-(amino-3,4-dioxo-1-cyclobuten-1-yl) amino]alkyl]-acid derivatives
US5157027A (en) 1991-05-13 1992-10-20 E. R. Squibb & Sons, Inc. Bisphosphonate squalene synthetase inhibitors and method
JP2648897B2 (en) * 1991-07-01 1997-09-03 塩野義製薬株式会社 Pyrimidine derivatives
FR2681784B1 (en) 1991-10-01 1995-06-09 Fabre Pierre Cosmetique DERMATOLOGICAL AND / OR COSMETOLOGICAL COMPOSITION CONTAINING RETINOUIDES AND USE OF NEW RETINOUIDES.
WO1993016388A1 (en) 1992-02-13 1993-08-19 The Uab Research Foundation Method of detecting and monitoring levels of 3'-amino-3'-deoxythymidine in body fluids and antibodies for same
GB2266527A (en) 1992-03-17 1993-11-03 Merck & Co Inc Substituted azetidinones useful in the treatment of leukemia
GB2266525A (en) 1992-03-17 1993-11-03 Merck & Co Inc Substituted cephalosporin sulfone compositions useful in the treatment of leukemia
GB9205917D0 (en) 1992-03-18 1992-04-29 Smithkline Beecham Plc Pharmaceuticals
DE69322087T2 (en) 1992-06-23 1999-04-08 Yamanouchi Pharma Co Ltd NEW HETEROCYCLIC BIS (PHOSPHONIC ACID) MONOHYDRATE DERIVATIVE CRYSTAL
US5532225A (en) 1992-07-31 1996-07-02 Sri International Acyclic purine phosphonate nucleotide analogs as antiviral agents, and related synthetic methods
US5358941A (en) 1992-12-02 1994-10-25 Merck & Co., Inc. Dry mix formulation for bisphosphonic acids with lactose
GB9301629D0 (en) 1993-01-27 1993-03-17 Scotia Holdings Plc Formulations containing unsaturated fatty acids
US5366965A (en) 1993-01-29 1994-11-22 Boehringer Mannheim Gmbh Regimen for treatment or prophylaxis of osteoporosis
SG47943A1 (en) 1993-02-10 1998-04-17 Astra Pharma Prod N-alkyl-2-substituted atp analogues
JP3526575B2 (en) 1993-03-08 2004-05-17 エーザイ株式会社 Phosphonic acid derivatives
EP0706387B1 (en) 1993-05-21 2004-08-18 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES New procedure to block the replication of reverse transcriptase dependent viruses by the use of inhibitors of deoxynucleotides synthesis
US5514798A (en) 1993-06-02 1996-05-07 Gilead Sciences, Inc. Method and cyclic carbonates for nucleotide analogues
DE69426904T2 (en) 1993-06-29 2001-10-11 Mitsubishi Chem Corp Phosphonate nucleotide ester derivatives
US5567689A (en) 1993-08-13 1996-10-22 The Uab Research Foundation Methods for increasing uridine levels with L-nucleosides
CA2637774C (en) 1993-09-10 2011-07-19 Emory University Nucleosides with anti-hepatitis b virus activity
FR2709754B1 (en) 1993-09-10 1995-12-01 Centre Nat Rech Scient Compounds 2 'or 3'-deoxy- and 2', 3'-dideoxy-beta-L-pentofuranonucleosides, preparation process and therapeutic application, in particular anti-viral.
US20020120130A1 (en) 1993-09-10 2002-08-29 Gilles Gosselin 2' or 3' -deoxy and 2', 3' -dideoxy-beta-L-pentofuranonucleo-side compounds, method of preparation and application in therapy, especially as anti- viral agents
EP0719273B1 (en) 1993-09-17 2010-11-03 Gilead Sciences, Inc. Nucleotide analogs
US5437772A (en) 1993-11-01 1995-08-01 The Electrosynthesis Co., Inc. Portable lead detector
US5567592A (en) 1994-02-02 1996-10-22 Regents Of The University Of California Screening method for the identification of bioenhancers through the inhibition of P-glycoprotein transport in the gut of a mammal
US5866679A (en) 1994-06-28 1999-02-02 Merck & Co., Inc. Peptides
US5599686A (en) 1994-06-28 1997-02-04 Merck & Co., Inc. Peptides
US6143864A (en) 1994-06-28 2000-11-07 Merck & Co., Inc. Peptides
WO1996001267A1 (en) 1994-07-04 1996-01-18 Takeda Chemical Industries, Ltd. Phosphonic acid compounds, their production and use
US5810856A (en) 1995-03-09 1998-09-22 Tveras; Rimvydas Wiping element for an oral hygiene device, window wiper, or the like
US5665713A (en) 1995-04-12 1997-09-09 Procter & Gamble Company Pharmaceutical composition for inhibiting the growth of viruses and cancers
US5665386A (en) 1995-06-07 1997-09-09 Avmax, Inc. Use of essential oils to increase bioavailability of oral pharmaceutical compounds
DE69636734T2 (en) 1995-06-07 2007-10-18 Emory University NUCLEOSIDE WITH ANTI-HEPATITIS B VIRUS EFFICACY
US5716928A (en) 1995-06-07 1998-02-10 Avmax, Inc. Use of essential oils to increase bioavailability of oral pharmaceutical compounds
US5627164A (en) 1995-06-13 1997-05-06 Exxon Research And Engineering Company Pyrazolyl borates complexes-(LAW294)
DE69630701T2 (en) 1995-07-17 2004-08-19 Cephalon, Inc. CYSTEIN AND SERINE PROTEASES INHIBITORS CONTAINING PHOSPHORUS
US5750493A (en) 1995-08-30 1998-05-12 Raymond F. Schinazi Method to improve the biological and antiviral activity of protease inhibitors
EP0876381A1 (en) 1995-12-19 1998-11-11 Darwin Discovery Limited Ifosfamide, analogues thereof and their preparation
CA2195262C (en) 1996-01-18 2005-08-09 Masaru Ubasawa Phosphonate nucleotide compounds
US5767097A (en) 1996-01-23 1998-06-16 Icn Pharmaceuticals, Inc. Specific modulation of Th1/Th2 cytokine expression by ribavirin in activated T-lymphocytes
JPH09241284A (en) 1996-03-08 1997-09-16 Koji Yamashita New oxyazaphosphorine derivative
US6128582A (en) 1996-04-30 2000-10-03 Vertex Pharmaceuticals Incorporated Molecules comprising an IMPDH-like binding pocket and encoded data storage medium capable of graphically displaying them
US5962440A (en) 1996-05-09 1999-10-05 Bristol-Myers Squibb Company Cyclic phosphonate ester inhibitors of microsomal triglyceride transfer protein and method
RU2111970C1 (en) 1996-06-25 1998-05-27 Иван Игоревич Федоров 3'-oximino-2',3'-dideoxynucleosides
GB9618634D0 (en) 1996-09-06 1996-10-16 Univ Southampton Anti cancer drugs
US6177404B1 (en) 1996-10-15 2001-01-23 Merck & Co., Inc. Conjugates useful in the treatment of benign prostatic hyperplasia
BR9712527A (en) 1996-10-16 2000-03-08 Icn Pharmaceuticals Monocyclic L-nucleosides, analogues and their uses
BR9713489A (en) 1996-10-24 2000-02-29 Novartis Ag substituted aminoalkanophosphonic acids
US5948750A (en) 1996-10-30 1999-09-07 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
GB9623908D0 (en) 1996-11-18 1997-01-08 Hoffmann La Roche Amino acid derivatives
PL333414A1 (en) 1996-11-19 1999-12-06 Icn Pharmaceuticals Multivalent salts of pyridostigmin and derivative compounds
SI20117A (en) 1996-12-27 2000-06-30 Icn Pharmaceuticals, Inc. G-rich oligo aptamers and methods of modulating an immune response
SE9700792L (en) 1997-03-06 1998-02-23 Carlo Castro Back lubricant for lubricating skin cream on a human body
AU6452098A (en) 1997-03-07 1998-09-22 Metabasis Therapeutics, Inc. Novel purine inhibitors of fructose-1,6-bisphosphatase
ATE253073T1 (en) 1997-03-07 2003-11-15 Metabasis Therapeutics Inc NEW BENZIMIDAZOLE INHIBITORS OF FRUCTOSE-1,6-BISPHOSPHATASE
AU6691798A (en) 1997-03-07 1998-09-22 Metabasis Therapeutics, Inc. Novel indole and azaindole inhibitors of fructose-1,6-bisphosphatase
GB9707659D0 (en) 1997-04-16 1997-06-04 Peptide Therapeutics Ltd Hepatitis C NS3 Protease inhibitors
BR9810364A (en) 1997-06-30 2000-09-05 Icn Pharmaceuticals Method for the production of thiazofurin and other c-nucleosides
US6130504A (en) 1997-07-11 2000-10-10 Sharp Kabushiki Kaisha Plasma addressing display device and method for producing the same
DE69826734T2 (en) 1997-07-25 2005-10-06 Gilead Sciences, Inc., Foster City Process for the preparation of adefovir dipivoxil
ES2234144T3 (en) 1997-08-11 2005-06-16 Boehringer Ingelheim (Canada) Ltd. ANALOGS OF INHIBITING PEPTIDES OF HEPATITIS C.
US6767991B1 (en) 1997-08-11 2004-07-27 Boehringer Ingelheim (Canada) Ltd. Hepatitis C inhibitor peptides
US5962522A (en) 1997-09-05 1999-10-05 Avmax, Inc. Propyl gallate to increase bioavailability of orally administered pharmaceutical compounds
US6180666B1 (en) 1997-09-05 2001-01-30 Anmax, Inc. Use of gallic acid esters to increase bioavailability of orally administered pharmaceutical compounds
US6391305B1 (en) 1997-09-10 2002-05-21 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
US20020115596A1 (en) 1997-10-27 2002-08-22 Merk & Co., Inc. Conjugates useful in the treatment of prostate cancer
US6423695B1 (en) 1998-01-13 2002-07-23 Ribapharm, Inc. Cytokine related treatments of disease
WO1999036074A1 (en) 1998-01-15 1999-07-22 Icn Pharmaceuticals, Inc. Use of 8-cl-camp in prevention of restenosis of arterial walls
US6348587B1 (en) 1998-02-25 2002-02-19 Emory University 2′-Fluoronucleosides
US6312662B1 (en) 1998-03-06 2001-11-06 Metabasis Therapeutics, Inc. Prodrugs phosphorus-containing compounds
ES2401070T3 (en) 1998-03-06 2013-04-16 Metabasis Therapeutics, Inc. New prodrugs for phosphorus-containing compounds
CA2289621A1 (en) 1998-03-16 1999-09-23 Ontogen Corporation Piperazines as inhibitors of fructose-1,6-bisphosphatase (fbpase)
GB9806815D0 (en) 1998-03-30 1998-05-27 Hoffmann La Roche Amino acid derivatives
WO1999050230A1 (en) 1998-03-31 1999-10-07 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly hepatitis c virus ns3 protease
MXPA00011473A (en) 1998-05-26 2002-04-24 Icn Pharmaceuticals Novel nucleosides having bicyclic sugar moiety.
GB9812523D0 (en) 1998-06-10 1998-08-05 Angeletti P Ist Richerche Bio Peptide inhibitors of hepatitis c virus ns3 protease
US6194391B1 (en) 1998-06-24 2001-02-27 Emory University 3′-azido-2′,3′-dideoxyuridine administration to treat HIV and related test protocol
IT1305313B1 (en) 1998-07-17 2001-05-04 Colla Paolo 3,4 - DIHYDRO- 6- BENZYL-4-OXOPYRIMIDINE REPLACED AND RELATED PROCESS OF PRODUCTION AND USE IN THE THERAPY OF HIV-1 INFECTIONS.
ES2531928T3 (en) 1998-08-10 2015-03-20 Novartis Ag Beta-L-2'-deoxy-nucleosides for the treatment of hepatitis B
US6444652B1 (en) 1998-08-10 2002-09-03 Novirio Pharmaceuticals Limited β-L-2'-deoxy-nucleosides for the treatment of hepatitis B
SK286080B6 (en) 1998-09-09 2008-03-05 Metabasis Therapeutics, Inc. Heterocyclic aromatic compounds possessing a phosphonate group, their use and pharmaceutical composition comprising the same
US6045638A (en) 1998-10-09 2000-04-04 Atlantic Research Corporation Monopropellant and propellant compositions including mono and polyaminoguanidine dinitrate
JP2002528499A (en) 1998-10-29 2002-09-03 ブリストル−マイヤーズ スクイブ カンパニー Amino nucleus-derived compounds that are inhibitors of the IMPDH enzyme
US6407077B1 (en) 1998-11-05 2002-06-18 Emory University β-L nucleosides for the treatment of HIV infection
MXPA01004504A (en) 1998-11-05 2005-04-19 Centre Nat Rech Scient Nucleosides with anti-hepatitis b virus activity.
US6756360B1 (en) 1998-12-24 2004-06-29 Metabasis Therapeutics, Inc. Combination of FBPase inhibitors and insulin sensitizers for the treatment of diabetes
US20020187945A1 (en) 1999-01-29 2002-12-12 Robert Tam Modulation of immune response by ribavirin
US7205404B1 (en) 1999-03-05 2007-04-17 Metabasis Therapeutics, Inc. Phosphorus-containing prodrugs
ES2272268T3 (en) 1999-03-05 2007-05-01 Metabasis Therapeutics, Inc. NEW PROFARMS THAT CONTAIN PHOSPHORUS.
US6608027B1 (en) 1999-04-06 2003-08-19 Boehringer Ingelheim (Canada) Ltd Macrocyclic peptides active against the hepatitis C virus
EP1196414B1 (en) 1999-06-25 2003-09-03 Vertex Pharmaceuticals Incorporated Prodrugs of carbamate inhibitors of impdh
PL354094A1 (en) 1999-08-27 2003-12-29 Icn Pharmaceuticals, Inc.Icn Pharmaceuticals, Inc. Pyrrolo[2,3-d]pyrimidine nucleoside analogs
EP1210354A1 (en) 1999-09-08 2002-06-05 Metabasis Therapeutics, Inc. Prodrugs for liver specific drug delivery
US6921763B2 (en) 1999-09-17 2005-07-26 Abbott Laboratories Pyrazolopyrimidines as therapeutic agents
EP1212327B8 (en) 1999-09-17 2004-02-25 Abbott GmbH & Co. KG Pyrazolopyrimidines as therapeutic agents
US6518253B1 (en) 1999-11-19 2003-02-11 Robert Tam Treatment of viral infections using the L-isomer of ribavirin
ES2300281T3 (en) 1999-12-03 2008-06-16 The Regents Of The University Of California At San Diego PHOSPHONATE COMPOUNDS.
AU5076601A (en) 1999-12-13 2001-07-03 Icn Pharmaceuticals, Inc. Pulmonary delivery of ribavirin or levovirinTM for systemic and quasi-systemic treatment of disease
JP2003523957A (en) 1999-12-23 2003-08-12 アイシーエヌ・ファーマシューティカルズ・インコーポレイテッド Compositions and methods for producing L-nucleosides, L-nucleotides and analogs thereof
US6770666B2 (en) 1999-12-27 2004-08-03 Japan Tobacco Inc. Fused-ring compounds and use thereof as drugs
AU3653301A (en) 2000-01-28 2001-08-07 Merck & Co., Inc. Treatment or prevention of prostate cancer with a cox-2 selective inhibiting drug
US6495677B1 (en) 2000-02-15 2002-12-17 Kanda S. Ramasamy Nucleoside compounds
US6455508B1 (en) 2000-02-15 2002-09-24 Kanda S. Ramasamy Methods for treating diseases with tirazole and pyrrolo-pyrimidine ribofuranosyl nucleosides
EP1259237A4 (en) 2000-02-17 2004-07-28 Merck & Co Inc Treatment or prevention of prostate cancer with a cox-2 selective inhibiting drug
WO2001077091A2 (en) 2000-04-05 2001-10-18 Tularik Inc. Ns5b hcv polymerase inhibitors
CN100457118C (en) 2000-04-13 2009-02-04 法玛塞特有限公司 3'-or 2'-hydroxymethyl substd. nucleoside derivs. for treatment of hepatitis virus infections
MY164523A (en) 2000-05-23 2017-12-29 Univ Degli Studi Cagliari Methods and compositions for treating hepatitis c virus
EP1736478B1 (en) 2000-05-26 2015-07-22 IDENIX Pharmaceuticals, Inc. Methods and compositions for treating flaviviruses and pestiviruses
EA005890B1 (en) 2000-05-26 2005-06-30 Айденикс (Кайман) Лимитед Methods for treating hepatitis delta virus infection with beta-l-2'-deoxy-nucleosides
US6875751B2 (en) 2000-06-15 2005-04-05 Idenix Pharmaceuticals, Inc. 3′-prodrugs of 2′-deoxy-β-L-nucleosides
GB0015627D0 (en) 2000-06-26 2000-08-16 Rademacher Group Limited Phosphoglycan messengers and their medical uses
US6448281B1 (en) 2000-07-06 2002-09-10 Boehringer Ingelheim (Canada) Ltd. Viral polymerase inhibitors
UA72612C2 (en) 2000-07-06 2005-03-15 Pyrido[2.3-d]pyrimidine and pyrimido[4.5-d]pyrimidine nucleoside analogues, prodrugs and method for inhibiting growth of neoplastic cells
DK1303527T3 (en) 2000-07-18 2005-01-31 Leo Pharma As matrix metalloprotease
GB0017676D0 (en) 2000-07-19 2000-09-06 Angeletti P Ist Richerche Bio Inhibitors of viral polymerase
PL213214B1 (en) 2000-07-21 2013-01-31 Gilead Sciences Prodrugs of phosphonate nucleotide analogues and methods for selecting and making same
US20030008841A1 (en) 2000-08-30 2003-01-09 Rene Devos Anti-HCV nucleoside derivatives
AU2001282528A1 (en) 2000-09-01 2002-03-22 Shionogi And Co., Ltd. Compounds having anti-hepatitis c virus effect
EP2251015B1 (en) 2000-10-18 2013-02-20 Gilead Pharmasset LLC Modified nucleosides for the treatment of viral infections and abnormal cellular proliferation
BR0115474A (en) 2000-11-17 2006-01-31 Idenix Cayman Ltd Composition and method for inhibiting hiv transmission using topically applied 6-benzyl-4-oxopyrimidines
US20040014696A1 (en) 2000-12-07 2004-01-22 Johnson Lau Specificity in treatment of diseases
KR100883184B1 (en) 2000-12-11 2009-02-12 암젠 인코포레이션 CXCR3 antagonists
IL155842A0 (en) 2000-12-12 2003-12-23 Schering Corp Diaryl peptides as ns3-serine protease inhibitors of hepatitis c virus
US6653295B2 (en) 2000-12-13 2003-11-25 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus NS3 protease
CA2429352A1 (en) 2000-12-15 2002-06-20 Lieven Stuyver Antiviral agents for treatment of flaviviridae infections
US20040063651A1 (en) 2000-12-26 2004-04-01 Masahiko Morioka Remedies for hepatitis c
MY134070A (en) 2001-01-22 2007-11-30 Isis Pharmaceuticals Inc Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
EP1390029B1 (en) 2001-04-11 2009-12-16 Idenix (Cayman) Limited Phenylindoles for the treatment of hiv
GB0114286D0 (en) 2001-06-12 2001-08-01 Hoffmann La Roche Nucleoside Derivatives
GB0119369D0 (en) 2001-08-08 2001-10-03 Bookham Technology Plc Optic system
GB2378524A (en) 2001-08-08 2003-02-12 Bookham Technology Plc Intergrated optic device
US20040077563A1 (en) 2001-08-21 2004-04-22 Johnson Lau Methods of drug delivery to hepatocytes and treatment of flaviviridae infections
US20030232760A1 (en) 2001-09-21 2003-12-18 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
EP1465667A4 (en) 2001-09-24 2007-06-27 Merck & Co Inc Screening and selection methods for statin drug combinations
EP1438054A4 (en) 2001-09-28 2006-07-26 Idenix Cayman Ltd Methods and compositions for treating flaviviruses and pestiviruses using 4'-modified nucleoside
US7138376B2 (en) 2001-09-28 2006-11-21 Idenix Pharmaceuticals, Inc. Methods and compositions for treating hepatitis C virus using 4'-modified nucleosides
WO2003037908A1 (en) 2001-10-31 2003-05-08 Ribapharm Inc. Antiviral combination therapy and compositions
AU2002353164A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Unusual nucleoside libraries, compounds, and preferred uses as antiviral and anticancer agents
AU2002349905A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Nucleoside libraries and compounds by mcc combinatorial strategies on solid support
AU2002359732A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Substituted purine nucleoside libraries and compounds by solid-phase combinatorial strategies
AU2002340387A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Cytidine libraries and compounds synthesized by solid-phase combinatorial strategies
AU2002357876A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Nucleoside analog libraries and compounds
AU2002353165A1 (en) 2001-12-17 2003-06-30 Ribapharm Inc. Deazapurine nucleoside libraries and compounds
AU2002359919A1 (en) 2001-12-28 2003-07-24 Terumo Kabushiki Kaisha Syringe
AU2002341942A1 (en) 2002-01-17 2003-09-02 Ribapharm Inc. Sugar modified nucleosides as viral replication inhibitors
CA2474563C (en) 2002-02-13 2010-11-09 Merck & Co., Inc. Methods of inhibiting orthopoxvirus replication with nucleoside compounds
IL163613A0 (en) 2002-02-19 2005-12-18 Cv Therapeutics Inc Partial and full agonists of a1 adenosine receptors
KR20050006221A (en) 2002-05-06 2005-01-15 제네랩스 테크놀로지스, 인코포레이티드 Nucleoside derivatives for treating hepatitis c virus infection
JP4476811B2 (en) 2002-05-13 2010-06-09 メタバシス・セラピューティクス・インコーポレイテッド New phosphonic acid prodrugs of PMEA and its analogs
CA2485597C (en) 2002-05-13 2013-07-02 Metabasis Therapeutics, Inc. Process for preparation of cyclic prodrugs of pmea and pmpa
US20060234962A1 (en) 2002-06-27 2006-10-19 Olsen David B Nucleoside derivatives as inhibitors of rna-dependent rna viral polymerase
US7608600B2 (en) 2002-06-28 2009-10-27 Idenix Pharmaceuticals, Inc. Modified 2′ and 3′-nucleoside prodrugs for treating Flaviviridae infections
KR20050035194A (en) 2002-06-28 2005-04-15 이데닉스 (케이만) 리미티드 2'-c-methyl-3'-o-l-valine ester ribofuranosyl cytidine for treatment of flaviviridae infections
US20040067877A1 (en) 2002-08-01 2004-04-08 Schinazi Raymond F. 2', 3'-Dideoxynucleoside analogues for the treatment or prevention of Flaviviridae infections
TW200418498A (en) 2002-09-30 2004-10-01 Genelabs Tech Inc Nucleoside derivatives for treating hepatitis C virus infection
US7094768B2 (en) 2002-09-30 2006-08-22 Genelabs Technologies, Inc. Nucleoside derivatives for treating hepatitis C virus infection
MXPA05004504A (en) 2002-10-31 2005-08-16 Metabasis Therapeutics Inc Novel cyclic phosphate diesters of 1,3-propane-1-aryl diols and their use in preparing prodrugs.
AU2003295684A1 (en) * 2002-11-15 2004-06-15 Teva Pharmaceutical Industries Ltd. Synthesis of 3,5-dihydroxy-7-pyrrol-1-yl heptanoic acids
BR0316363A (en) 2002-11-15 2005-10-04 Idenix Cayman Ltd 2'-branched nucleosides and flaviviridae mutation
TWI332507B (en) 2002-11-19 2010-11-01 Hoffmann La Roche Antiviral nucleoside derivatives
TWI294882B (en) 2002-12-09 2008-03-21 Hoffmann La Roche Anhydrous crystalline azido cytosine hemisulfate derivative
PL3521297T3 (en) 2003-05-30 2022-04-04 Gilead Pharmasset Llc Modified fluorinated nucleoside analogues
PL1658302T3 (en) 2003-07-25 2011-03-31 Idenix Pharmaceuticals Inc Purine nucleoside analogues for treating diseases caused by flaviviridae including hepatitis c
TW200510425A (en) 2003-08-13 2005-03-16 Japan Tobacco Inc Nitrogen-containing fused ring compound and use thereof as HIV integrase inhibitor
US7491794B2 (en) 2003-10-14 2009-02-17 Intermune, Inc. Macrocyclic compounds as inhibitors of viral replication
US7144868B2 (en) 2003-10-27 2006-12-05 Genelabs Technologies, Inc. Nucleoside compounds for treating viral infections
KR20060096487A (en) 2003-10-27 2006-09-11 진랩스 테크놀러지스, 인크. Nucleoside compounds for treating viral infections
TW200517106A (en) 2003-10-29 2005-06-01 Wyeth Corp Sustained release pharmaceutical compositions
US20060014740A1 (en) 2003-11-18 2006-01-19 Miller Duane D Analogs exhibiting inhibition of cell proliferation, methods of making, and uses thereof
JPWO2005063222A1 (en) 2003-12-26 2007-07-19 協和醗酵工業株式会社 Hsp90 family protein inhibitors
EP2204374B1 (en) 2003-12-30 2012-06-13 Gilead Sciences, Inc. Nucleoside phosphonates and analogs thereof for the treatment of HPV-infections
US20050182252A1 (en) 2004-02-13 2005-08-18 Reddy K. R. Novel 2'-C-methyl nucleoside derivatives
US20090275099A1 (en) 2004-04-27 2009-11-05 Regents Of The University Of Michigan Methods and compositions for treating diseases and conditions associated with mitochondrial function
US7582758B2 (en) 2004-06-08 2009-09-01 Metabasis Therapeutics, Inc. Lewis acid mediated synthesis of cyclic esters
US7206468B2 (en) 2004-08-17 2007-04-17 Huang Hung-Chia Broad-band fiber-optic wave plates
US20060270634A1 (en) 2005-05-06 2006-11-30 Miller Duane D Acetal phosphate-derived LPA mimics, PPARgamma activators, and autotaxin inhibitors
RU2414475C2 (en) 2005-06-29 2011-03-20 Трешолд Фармасьютикалз, Инк. Phosphoramidate alkylator prodrug
JP2009502790A (en) 2005-07-19 2009-01-29 ザ ユニバーシティー オブ テネシー リサーチ ファウンデーション CFTR LPA2 receptor agonist inhibitor
WO2007020193A2 (en) 2005-08-15 2007-02-22 F. Hoffmann-La Roche Ag Antiviral phosphoramidates of 4 ' -substituted pronucleotides
JP4954573B2 (en) 2006-02-28 2012-06-20 オリンパス株式会社 Endoscope system
EP1834944A1 (en) * 2006-03-17 2007-09-19 Ratiopharm GmbH Process for preparing C7 intermediates and their use in the preparation on N-substituted pyrrole derivatives
US7795291B2 (en) 2006-07-07 2010-09-14 Bristol-Myers Squibb Company Substituted acid derivatives useful as anti-atherosclerotic, anti-dyslipidemic, anti-diabetic and anti-obesity agents and method
WO2009011850A2 (en) 2007-07-16 2009-01-22 Abbott Laboratories Novel therapeutic compounds
ES2618482T3 (en) 2007-08-10 2017-06-21 Basil Rigas Anti-inflammatory compounds and uses thereof
WO2009073506A2 (en) 2007-11-29 2009-06-11 Metabasis Therapeutics Inc. Nucleoside prodrugs and uses thereof
AR069740A1 (en) 2007-11-29 2010-02-17 Metabasis Therapeutics Inc ANTIVIRAL NUCLEOSID COMPOUNDS
IT1391812B1 (en) 2008-07-29 2012-01-27 Maria Grazia Rimoli KETOROLAC GALACTOSILATE PROFARM FOR ANALGESIC AND ANTI-INFLAMMATORY USE WITH BETTER PHARMACOKINETIC CHARACTERISTICS AND TOXICOLOGICAL PROFILE OF THE STARTING DRUG
WO2010042600A2 (en) 2008-10-08 2010-04-15 The Uab Research Foundation Photo-activatable therapeutic agents and methods of using
EP2351762B1 (en) * 2008-10-20 2013-10-16 Kaneka Corporation NOVEL PYRIMIDINE DERIVATIVE AND METHOD FOR PRODUCING HMG-CoA REDUCTASE INHIBITOR INTERMEDIATE
EP2370438A1 (en) 2008-12-04 2011-10-05 Exelixis, Inc. Imidazo [1,2a]pyridine derivatives, their use as s1p1 agonists and methods for their production
WO2010105048A1 (en) 2009-03-12 2010-09-16 Metabolic Solutions Development Company Thiazolidinedione analogues
EP2493905B1 (en) 2009-10-26 2016-11-30 Warner Chilcott Company, LLC Bisphosphonate compounds for treating bone metabolism disorders
AU2010340087A1 (en) 2009-12-21 2012-07-26 Merck Sharp & Dohme Corp. Tyrosine kinase inhibitors
WO2011160974A2 (en) * 2010-06-21 2011-12-29 Nicox S.A. Statin derivatives
KR101292238B1 (en) * 2010-07-01 2013-07-31 주식회사유한양행 Process for the preparation of HMG-CoA reductase inhibitors and intermediates thereof
MX2013013570A (en) 2011-05-19 2014-07-09 Univ Emory Purine monophosphate prodrugs for treatment of viral infections.
SG195018A1 (en) 2011-06-06 2013-12-30 Arbor Therapeutics Llc Acid-labile lipophilic prodrugs of cancer chemotherapeutic agents
EP2780026B1 (en) 2011-11-15 2019-10-23 Merck Sharp & Dohme Corp. Hcv ns3 protease inhibitors
MA37519B1 (en) 2012-05-15 2017-03-31 Novartis Ag Compounds and compositions for inhibiting the activity of abl1, abl2 and bcr-abl1
US20140142052A1 (en) 2012-05-17 2014-05-22 Normoxys, Inc. Phosphorylated polyols, pyrophosphates, and derivatives thereof having biological activity
US9353061B2 (en) * 2012-07-20 2016-05-31 Academia Sinica 3,5,N-trihydroxy-alkanamide and derivatives: method for making same and use thereof
US9326991B2 (en) 2012-09-14 2016-05-03 Ligand Pharmaceuticals, Inc. Nucleotide prodrug compounds and use
EP2978744A1 (en) * 2013-03-29 2016-02-03 DSM Sinochem Pharmaceuticals Netherlands B.V. Amine salts of pitavastatin and rosuvastatin
CN106061984A (en) 2014-02-13 2016-10-26 配体药物公司 Prodrug compounds and their uses
WO2016003812A1 (en) 2014-07-02 2016-01-07 Ligand Pharmaceuticals, Inc. Prodrug compounds and uses therof
WO2016130417A1 (en) 2015-02-11 2016-08-18 Omthera Pharmaceuticals Inc Omega-3 fatty acid prodrug compounds and uses thereof
BR112020024491A2 (en) * 2018-06-05 2021-03-02 Novozymes Bioag A/S methods of foliar application of a composition, of controlling plant pests on a plant or part of a plant and / or inducing resistance to a plant pest on a plant or part of a plant, of controlling or preventing pest damage in a plant plant propagation material, a plant, part of a plant and / or plant organ, to control or prevent damage from a lepidopteran pest on a plant and to control or prevent damage from an insect pest on a plant.

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987005297A1 (en) 1986-03-03 1987-09-11 The University Of Chicago Cephalosporin derivatives
US8207333B2 (en) * 2005-06-24 2012-06-26 Lek Pharmaceuticals D. D. Process for preparing pure amorphous rosuvastatin calcuim
US8354530B2 (en) * 2005-07-28 2013-01-15 Lek Pharmaceuticals d. d Process for the synthesis of rosuvastatin calcium
WO2007125547A2 (en) * 2006-05-03 2007-11-08 Manne Satyanarayana Reddy Novel process for statins and its pharmaceutically acceptable salts thereof
KR20100087931A (en) * 2009-01-29 2010-08-06 (주)유케이케미팜 Precursive compound of rosuvastatin and manufacturing method for the precursive compound of rosuvastatin
WO2010103320A1 (en) * 2009-03-10 2010-09-16 Bradford Pharma Limited Use of rosuvastatin lactols as medicaments
CN102079726A (en) * 2009-11-27 2011-06-01 上海医药工业研究院 Miazine compounds, intermediates of miazine compounds, preparation method of intermediates and miazine compounds as well as application of miazine compound
WO2012038785A1 (en) * 2010-09-21 2012-03-29 Biocon Limited Polymorphs of rosuvastatin acetonide calcium ((3r,5s,6e)-7-[4-(4- fluorophenyl)-6-isopropyl-2-(methanesulfonyl-methyl-amino)-pyrimn)in-5- yl)vinyl)-2,2-dimethyl-l,3-dioxan-4-yl) acetic acid calcium salt
US20170183314A1 (en) * 2013-11-25 2017-06-29 Fudan University Method for preparing rosuvastatin sodium
KR20160126700A (en) * 2015-04-24 2016-11-02 미래파인켐 주식회사 New Statin intermediate, the preparation of the same and the preparation of Rosuvastatin using the same
KR20170078033A (en) * 2015-12-29 2017-07-07 미래파인켐 주식회사 Novel Statin intermediate, the preparation method of the same and the preparation method of Rosuvastatin using the same
WO2017137469A1 (en) * 2016-02-11 2017-08-17 Stichting Katholieke Universiteit Novel class of compounds for the treatment of cardiovascular disease
CN107382875A (en) * 2017-06-26 2017-11-24 浙江美诺华药物化学有限公司 A kind of synthetic method of rosuvastain calcium chiral isomer impurity

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
"Encyclopedia of Reagents for Organic Synthesis", 1995, JOHN WILEY AND SONS
"Goodman and Gilman's: The Pharmacological Basis of Therapeutics", 1990, PERGAMON PRESS
"Modern Pharmaceutics", 2002
"Protective Groups in Organic Chemistry", 1973, PLENUM PRESS
"Remington's The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
AHMAD, S. ET AL.: "3R,5S,E)-7-(4-(4-Fluorophenyl)-6-isopropyl-2-(methyl(1-methyl- 1H-1,2,4-triazol-5-yl)amino)pyrimidin-5-yl)-3,5-dihydroxyhept-6-enoic acid (BMS- 644950): A rationally designed orally efficacious 3-hydroxy-3-methylglutaryl coenzyme-A reductase inhibitor with reduced myotoxicity potential", JOURNAL OF MEDICINAL CHEMISTRY, vol. 51, 2008, pages 2722 - 2733, XP055003825, doi:10.1021/jm800001n *
ANSEL: "Introduction to Pharmaceutical Dosage Forms", 2004
LEE, Y. H. ET AL.: "Synthesis and characterization of rosuvastatin calcium impurity A; a HMG-CoA reductase inhibitor", TETRAHEDRON LETTERS, vol. 58, no. 26, 28 February 2017 (2017-02-28), pages 2614 - 2617, XP055625067 *
LIEBERMAN ET AL., PHARMACEUTICAL DOSAGE FORMS: TABLETS, 1989
NEMA ET AL.: "Excipients and Their Role in Approved Injectable Products: Current Usage and Future Directions", PDA J PHARM SCI AND TECH, vol. 65, 2011, pages 287 - 332, XP009166667, DOI: 10.5731/pdajpst.2011.00634
P.G.M. GREENT.W. WUTTS: "Protecting Groups in Organic Synthesis", 1999, WILEY
POWELL ET AL.: "Compendium of Excipients for Parenteral Formulations", PDA J PHARM SCI AND TECH, vol. 52, 1998, pages 238 - 311, XP009119027
See also references of EP3737676A4

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4005633A2 (en) 2022-02-08 2022-06-01 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester
EP4005633A3 (en) * 2022-02-08 2022-06-08 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester
WO2023152754A1 (en) * 2022-02-08 2023-08-17 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester
WO2023152761A1 (en) * 2022-02-08 2023-08-17 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester

Also Published As

Publication number Publication date
EP3737676B1 (en) 2024-03-06
US20200339551A1 (en) 2020-10-29
EP3737676A1 (en) 2020-11-18
US11970482B2 (en) 2024-04-30
RU2020126177A (en) 2022-02-10
CN111788196A (en) 2020-10-16
CA3087932A1 (en) 2019-07-18
JP2021509907A (en) 2021-04-08
EP3737676A4 (en) 2021-09-15
KR20200123106A (en) 2020-10-28
AU2019207625A1 (en) 2020-07-30

Similar Documents

Publication Publication Date Title
WO2019139919A1 (en) Acetal compounds and therapeutic uses thereof
WO2020172615A1 (en) Compounds for the treatment of ocular disease
CN113767103A (en) Novel spirocyclic K-Ras G12C inhibitor
AU2016291708A1 (en) Plinabulin compositions
KR20010031010A (en) (Benzodioxan, benzofuran or benzopyran) derivatives having fundic relaxation properties
ES2901401T3 (en) Dihydropyrimidine compound and method of preparation and use thereof
US20020065261A1 (en) Cryptophycin compound
US10633399B2 (en) Functionalized aminobenzoboroxoles
BR112020025701A2 (en) PRODUCTION PROCESS AND INTERMEDIARIES FOR A PIRROLEUM COMPOUND [2,3-D] PYRIMIDINE AND ITS USE
KR101765956B1 (en) Method of preparing (+)-1,4-dihydro-7-[(3s,4s)-3-methoxy-4-(methylamino)-1-pyrrolidinyl]-4-oxo-1-(2-thiazolyl)-1,8-naphthyridine-3-carboxylic acid
US7795281B2 (en) Optically active dihydropyridine derivative
WO2022214937A1 (en) Substituted tetracyclic carboxylic acids, analogues thereof, and methods using same
CN115443132A (en) Controlled delivery of chromancarlin prodrugs
WO2019042443A1 (en) Compound having tyrosine protein kinase jak3-degradation activity
US20230019280A1 (en) Substituted isoindolonyl 2,2'-bipyrimidinyl compounds, analogues thereof, and methods using same
RU2773843C1 (en) Plinabulin composites
US11166954B2 (en) Dihydropyrimidine compound and preparation method and use thereof
US20230312481A1 (en) Substituted (phthalazin-1-ylmethyl)ureas, substituted n-(phthalazin-1-ylmethyl)amides, and analogues thereof
ES2262515T3 (en) R-HYDROXINEPHAZODONE.
US20180282297A1 (en) Polymorphs of 3-(4-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione
WO2023021143A1 (en) Quinazoline-thiohydantoin fused heterocycles which are suitable for treating, ameliorating or preventing a proliferative disorder
AU2022313576A1 (en) Synthesis of substituted tricyclic amides and analogues thereof
KR20230114274A (en) Prodrugs of 5α-hydroxy-6β-[2-(1H-imidazol-4-yl)ethylamino]cholestan-3β-ol and pharmaceutical compositions comprising them for use in cancer treatment
WO2023235480A1 (en) Aryl hydantoin compounds and methods of use
WO2018224419A1 (en) Nitric oxide donating isomannide derivatives

Legal Events

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

Ref document number: 19738393

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
ENP Entry into the national phase

Ref document number: 3087932

Country of ref document: CA

Ref document number: 2020537621

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019207625

Country of ref document: AU

Date of ref document: 20190108

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019738393

Country of ref document: EP

Effective date: 20200810