WO2016141890A1 - Composés en tant qu'inhibiteurs du virus de l'hépatite c et utilisations pharmaceutiques associées - Google Patents

Composés en tant qu'inhibiteurs du virus de l'hépatite c et utilisations pharmaceutiques associées Download PDF

Info

Publication number
WO2016141890A1
WO2016141890A1 PCT/CN2016/076131 CN2016076131W WO2016141890A1 WO 2016141890 A1 WO2016141890 A1 WO 2016141890A1 CN 2016076131 W CN2016076131 W CN 2016076131W WO 2016141890 A1 WO2016141890 A1 WO 2016141890A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
alkoxy
heterocyclyl
heteroaryl
Prior art date
Application number
PCT/CN2016/076131
Other languages
English (en)
Inventor
Yingjun Zhang
Hongming XIE
Qinghong Fang
Zhiqiang Liu
Bailin HU
Jiancun Zhang
Original Assignee
Sunshine Lake Pharma Co., Ltd.
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 Sunshine Lake Pharma Co., Ltd. filed Critical Sunshine Lake Pharma Co., Ltd.
Publication of WO2016141890A1 publication Critical patent/WO2016141890A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/113Spiro-condensed systems with two or more oxygen atoms as ring hetero atoms in the oxygen-containing ring

Definitions

  • the present invention belongs to pharmaceutical field, specificially, it relates to compounds and compositions thereof for treating Hepatitis C virus (HCV) infection, uses thereof, and application methods thereof.
  • HCV Hepatitis C virus
  • HCV is a major human pathogen, infecting an estimated 170 million persons worldwide —roughly five times the number infected by human immunodeficiency virus type 1. A substantial fraction of these HCV infected individuals develop serious progressive liver disease, including cirrhosis and hepatocellular carcinoma. Chronic HCV infection is thus a major worldwide cause of liver-related premature mortality.
  • HCV is a positive-stranded RNA virus. Based on a comparison of the deduced amino acid sequence and the extensive similarity in the 5’untranslated region, HCV has been classified as a separate genus in the Flaviviridae family. All members of the Flaviviridae family have enveloped virions that contain a positive stranded RNA genome encoding all known virus-specific proteins via translation of a single, uninterrupted, open reading frame (ORF) .
  • ORF open reading frame
  • HCV infected cells viral RNA is translated into a polyprotein that is cleaved into ten individual proteins. At the amino terminus are structural proteins, followed by E1 and E2. Additionally, there are six non-structural proteins (NS2, NS3, NS4A, NS4B, NS5A and NS5B) , which play a significant role in the HCV lifecycle (see, for example, Lindenbach et al., Nature, 2005, 436, 933-938) .
  • HCV human immunodeficiency virus
  • the single strand HCV RNA genome is approximately 9500 nucleotides in length and has a single open reading frame (ORF) encoding a single large polyprotein of about 3000 amino acids. In infected cells, this polyprotein is cleaved at multiple sites by cellular and viral proteases to produce the structural and non-structural (NS) proteins. In the case of HCV, the generation of mature non-structural proteins (NS2, NS3, NS4A, NS4B, NS5A and NS5B) is effected by two viral proteases.
  • ORF open reading frame
  • the first one is believed to be a metalloprotease and cleaves at the NS2-NS3 junction; the second one is a serine protease within the N-terminal region of NS3 (also referred herein as NS3 protease) and mediates all the subsequent cleavages downstream of NS3, both in cis, at the NS3-NS4A cleavage site, and in trans, at the remaining NS4A-NS4B, NS4B-NS5A, NS5A-NS5B sites.
  • the NS4A protein appears to serve multiple functions, acting as a cofactor for the NS3 protease and possibly assisting in the membrane localization of NS3 and other viral replicase components.
  • NS5B (also referred to herein as HCV polymerase) is a RNA-dependent RNA polymerase that is involved in the replication of HCV.
  • the compounds disclosed herein are used for treating HCV-infected patients, and which selectively inhibit HCV viral replication.
  • the present invention refers to a method of anti HCV infection.
  • the compounds or composition thereof provided herein have good inhibitory effect on HCV GT1a, GT1b, GT2a, GT3a, GT4b, GT5a, GT6a, and HCVGT1b L31V, GT1b Y93H resistant strains. Therefore, the invention provides durgs as genome-wide HCV inhibitors, which have anti drug-resistance effect and good bioavailability.
  • a compound having Formula (I) or a stereoisomer a geometric isomer, a tautomer, an enantiomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof,
  • each of X and X 1 is independently N or CR 7b ;
  • each of R 1 , R 2 , R 3 and R 4 is independently H, deuterium, alkyl, arylalkyl, cycloalkyl, heterocyclyl, heteroaryl or aryl; or R 1 and R 2 , together with the X-CH to which they are attached, optionally form a 3-8 membered heterocyclic ring, 3-8 membered carbocyclic ring, fused bicyclic ring, fused heterobicyclic ring, spiro bicyclic ring or spiro heterobicyclic ring; or R 3 and R 4 , together with the X 1 -CH to which they are attached, optionally form a 3-8 membered heterocyclic ring, 3-8 membered carbocyclic ring, fused bicyclic ring, fused heterobicyclic ring, spiro bicyclic ring or spiro heterobicyclic ring;
  • each f, n and t is independently 0, 1, 2, 3 or 4;
  • n 1, 2, 3 or 4;
  • each r is independently 0, 1 or 2;
  • each alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, arylalkyl, -C ( O) - (CR 8 R 8a ) t -N (R 9 ) -R 10 , ⁇ -amino acid group, 3-8 membered heterocyclic ring, 3-8 membered carbocyclic ring, fused bicyclic ring, fused heterobicyclic ring, spiro bicyclic ring, spiro heterobicyclic ring, haloalkyl, alkenyl, alkynyl, alkoxyalkyl, heteroarylalkyl, cycloalkylalkyl, heterocyclylalkyl, arylamino, heteroarylamino, arylalkylamino, heteroarylalkylamino, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, heterocyclyloxy, heterocyclyl
  • each of R 1 , R 2 , R 3 and R 4 is independently H, C 1-6 alkyl, C 6-10 aryl-C 1-6 -alkyl, C 3-10 cycloalkyl, C 2-10 heterocyclyl, C 1-9 heteroaryl or C 6-10 aryl; or R 1 and R 2 , together with the X-CH to which they are attached, optionally form a 3-8 membered heterocyclic ring, 3-8 membered carbocyclic ring, C 5-12 fused bicyclic ring, C 5-12 fused heterobicyclic ring, C 5-12 spiro bicyclic ring or C 5-12 spiro heterobicyclic ring; or R 3 and R 4 , together with the X 1 -CH to which they are attached, optionally form a 3-8 membered heterocyclic ring, 3-8 membered carbocyclylic ring, C 5-12 fused bicyclic ring, C 5-12 fused heterobicyclic ring
  • R 1 and R 2 together with Y-X-CH to which they are attached, form one of the following groups:
  • R 3 and R 4 together with Y 1 -X 1 -CH to which they are attached, form one of the following groups:
  • each R 9b is independently H, deuterium, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, C 1-6 alkoxy-C 1-6 -alkyl, C 1-6 alkylamino-C 1-6 -alkyl, C 1-6 alkylthio-C 1-6 -alkyl, C 6-10 aryl-C 1-6 -alkyl, C 1-9 heteroaryl, C 6-10 aryl, C 2-10 heterocyclyl or C 3-8 cycloalkyl; and
  • each n 1 and n 2 is independently 1, 2, 3 or 4.
  • each R 9b is independently H, deuterium, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 aminoalkyl, C 1-3 alkoxy-C 1-3 -alkyl, C 1-3 alkylamino-C 1-3 -alkyl, C 1-3 alkylthio-C 1-3 -alkyl, C 6-10 aryl-C 1-3 -alkyl, C 1-9 heteroaryl, C 6-10 aryl, C 2-10 heterocyclyl or C 3-8 cycloalkyl.
  • each R 9b is independently H, deuterium, methyl, ethyl, propyl, isopropyl, trifluoromethyl, hydroxymethyl, aminomethyl, methoxymethyl, ethoxymethyl, phenylmethyl, phenyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
  • each r is independently 0, 1 or 2.
  • each R 8 and R 8a is independently H, deuterium, hydroxy, amino, F, Cl, Br, I, cyano, C 1-6 alkyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 2-10 heterocyclyl, C 3-8 cycloalkyl, mercapto, nitro, C 6-10 aryl-C 1-6 -alkyl, C 1-6 alkoxy-C 1-6 -alkyl, C 1-9 heteroaryl-C 1-6 -alkyl, C 3-8 cycloalkyl-C 1-6 -alkyl, C 2-10 heterocyclyl-C 1-6 -alkyl, C 6-10 arylamino, C 1-9 heteroarylamino, C 6-10 aryl-C 1-6 -alkylamino, C 1-9 heteroaryl-C 1-6 -alkylamino, C 1-9 heteroaryloxy
  • t 0, 1, 2, 3 or 4;
  • each r is independently 0, 1 or 2.
  • the compound of the invention has Formula (II) , (IIa) , (IIb) or (III) , or a stereoisomer, a geometric isomer, a tautomer, an enantiomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof,
  • A is -O-, -S-, -NH-, -CH 2 -S-, -CH 2 -O-, -CH 2 -NH-, -O-CH 2 -, -NH-CH 2 -, -S-CH 2 -or -CH 2 -CH 2 -;
  • each n 1 and n 2 is independently 1, 2, 3 or 4.
  • each R 8 and R 8a is independently H, deuterium, hydroxy, amino, F, Cl, Br, I, cyano, C 1-6 alkyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 2-10 heterocyclyl, C 3-8 cycloalkyl, mercapto, nitro, C 6-10 aryl-C 1-6 -alkyl, C 1-6 alkoxy-C 1-6 -alkyl, C 1-9 heteroaryl-C 1-6 -alkyl, C 3-8 cycloalkyl-C 1-6 -alkyl, C 2-10 heterocyclyl-C 1-6 -alkyl, C 6-10 arylamino, C 1-9 heteroarylamino, C 6-10 aryl-C 1-6 -alkylamino, C 1-9 heteroaryl-C 1-6 -alkylamino, C 1-9 heteroaryl-
  • each R 8 and R 8a is independently H, deuterium, hydroxy, amino, F, Cl, Br, I, cyano, methyl, ethyl, isopropyl, n-butyl, i-butyl, s-butyl, t-butyl, deuterated methyl, deuterated ethyl, deuterated propyl, deuterated isopropoxy, methoxy, ethoxy, isopropoxy, methoxymethyl, 1-methoxyethyl, 2-methoxyethyl, 1-methoxypropyl, 2-methoxypropyl, 3-methoxypropyl, phenyl, pyranyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, trifluoromethyl, vinyl, allyl, ethynyl, morpholinyl, mercapto, nitro, phenylmethyl or phenylamin
  • composition comprising any one of the compounds described above.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle or a combination thereof.
  • the pharmaceutical composition further comprises an anti-HCV agent other than the said compound, wherein the anti-HCV agent is interferon, ribavirin, IL-2, IL-6, IL-12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, imiquimod, an inosine 5’-monophosphate dehydrogenase inhibitor, amantadine, rimantadine, bavituximab, hepatitis C immunogloblin, Civacir TM , boceprevir, telaprevir, erlotinib, daclatasvir, simeprevir, asunaprevir, vaniprevir, faldaprevir, paritaprevir, danoprevir, sovaprevir, grazoprevir, vedroprevir, BZF-961, GS-9256, narlaprevir, ANA975, ombitasvir, EDP239,
  • the anti-HCV agent is used for inhibiting HCV replication process and/or at least a function of a HCV viral protein; and wherein the HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress; and wherein the HCV viral protein comprises metalloproteinase, non-structural protein NS2, NS3, NS4A, NS4B, NS5A or NS5B, an internal ribosome entry site (IRES) and inosine-5’-monophosphate dehydrogenase (IMPDH) required in the HCV viral replication.
  • the HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress
  • the HCV viral protein comprises metalloproteinase, non-structural protein NS2, NS3, NS4A, NS4B, NS5A or NS5B, an internal ribosome entry site (IRES) and inosine-5
  • HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress; and wherein the HCV viral protein comprises metalloproteinase, non-structural protein NS2, NS3, NS4A, NS4B, NS5A or NS5B, an internal ribosome entry site (IRES) and inosine-5’-monophosphate dehydrogenase (IMPDH) required in the HCV viral replication.
  • IRS internal ribosome entry site
  • IMPDH inosine-5’-monophosphate dehydrogenase
  • provided herein is use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for preventing, managing, treating or lessening the severity of HCV infection or a HCV disorder in a subject.
  • the compound or the pharmaceutical composition disclosed herein for use in inhibiting HCV replication process and/or at least a function of a HCV viral protein; wherein the HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress; and wherein the HCV viral protein comprises metalloproteinase, non-structural protein NS2, NS3, NS4A, NS4B, NS5A or NS5B, an internal ribosome entry site (IRES) and inosine-5’-monophosphate dehydrogenase (IMPDH) required in the HCV viral replication.
  • the HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress
  • the HCV viral protein comprises metalloproteinase, non-structural protein NS2, NS3, NS4A, NS4B, NS5A or NS5B, an internal ribosome entry site (IRES) and inos
  • provided herein is the compound or the pharmaceutical composition disclosed herein for use in preventing, managing, treating or lessening the severity of HCV infection or a HCV disorder in a subject.
  • a method of inhibiting HCV replication process and/or at least a function of a HCV viral protein in a subject comprising administering to the subject the compound or the pharmaceutical composition disclosed herein; wherein the HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress; and wherein the HCV viral protein comprises metalloproteinase, non-structural protein NS2, NS3, NS4A, NS4B, NS5A or NS5B, an internal ribosome entry site (IRES) and inosine-5’-monophosphate dehydrogenase (IMPDH) required in the HCV viral replication.
  • the HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress
  • the HCV viral protein comprises metalloproteinase, non-structural protein NS2, NS3, NS4A, NS4B, NS5A or NS5B, an internal rib
  • provided herein is a method of preventing, managing, treating or lessening the severity of HCV infection or a HCV disorder in a subject, comprising administering to the subject the compound or the pharmaceutical composition disclosed herein.
  • provided herein is a method of preparing, separating or purifying the compound of Formula (I) , (II) , (IIa) , (IIb) or (III) .
  • grammatical articles “a” , “an” and “the” are intended to include “at least one” or “one or more” unless otherwise indicated herein or clearly contradicted by the context.
  • the articles are used herein to refer to one or more than one (i.e. at least one) of the grammatical objects of the article.
  • a component means one or more components, and thus, possibly, more than one component is contemplated and may be employed or used in an implementation of the described embodiments.
  • the term “subject” refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans, male or female) , cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human.
  • primates e.g., humans, male or female
  • the subject is a primate.
  • the subject is a human.
  • patient refers to a human (including adults and children) or other animal. In one embodiment, “patient” refers to a human.
  • Stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. Stereoisomers include enantiomer, diastereomers, conformer (rotamer) , geometric (cis/trans) isomer, atropisomer, etc.
  • Chiral refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • Enantiomers refers to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boling points, spectral properties or biological activities. Mixture of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography such as HPLC.
  • optically active compounds Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light.
  • the prefixes D and L, or R and S are used to denote the absolute configuration of the molecule about its chiral center (s) .
  • the prefixes d and l or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or l meaning that the compound is levorotatory.
  • a compound prefixed with (+) or d is dextrorotatory.
  • a specific stereoisomer may be referred to as an enantiomer, and a mixture of such stereoisomers is called an enantiomeric mixture.
  • a 50: 50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
  • any asymmetric atom (e.g., carbon or the like) of the compound (s) disclosed herein can be present in racemic or enantiomerically enriched, for example the (R) -, (S) -or (R, S) -configuration.
  • each asymmetric atom has at least 50 %enantiomeric excess, at least 60 %enantiomeric excess, at least 70 %enantiomeric excess, at least 80 %enantiomeric excess, at least 90 %enantiomeric excess, at least 95 %enantiomeric excess, or at least 99 %enantiomeric excess in the (R) -or (S) -configuration.
  • the compounds can be present in the form of one of the possible stereoisomers or as mixtures thereof, such as racemates and diastereoisomer mixtures, depending on the number of asymmetric carbon atoms.
  • Optically active (R) -and (S) -isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituent may be E or Z configuration. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis-or trans-configuration.
  • Any resulting mixtures of stereoisomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric isomers, enantiomers, diastereomers, for example, by chromatography and/or fractional crystallization.
  • racemates of final products or intermediates can be resolved into the optical antipodes by methods known to those skilled in the art, e.g., by separation of the diastereomeric salts thereof.
  • Racemic products can also be resolved by chiral chromatography, e.g., high performance liquid chromatography (HPLC) using a chiral adsorbent.
  • HPLC high performance liquid chromatography
  • Preferred enantiomers can also be prepared by asymmetric syntheses. See, for example, Jacques, et al., Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981) ; Principles of Asymmetric Synthesis (2nd Ed. Robert E.
  • tautomer or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier. Where tautomerization is possible (e.g. in solution) , a chemical equilibrium of tautomers can be reached.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • keto-enol tautomerization is the interconversion of pentane-2, 4-dione and 4-hydroxypent-3-en-2-one tautomers.
  • tautomerization is phenol-keto tautomerization.
  • the specific example of phenol-keto tautomerisms is pyridin-4-ol and pyridin-4 (1H) -one tautomerism. Unless otherwise stated, all tautomeric forms of the compounds disclosed herein are within the scope of the invention.
  • Annular tautomerism is a type of prototropic tautomerism wherein a proton can occupy two or more positions of a heterocyclic system. Both isomers coexsit in a balanced system at a relatively high rate of conversion to each other, for example, 1H-and 3H-imidazole; 1H-, 2H-and 4H-1, 2, 4-triazole; 1H-and 2H-isoindole.
  • the structural fragments refered herein, for example, Aa and Ab, or, Ba and Bb are annular tautomers.
  • the invention only describes the structure of one of the annular tautomers at anywhere, that denotes the invention also describes the other structure of the annular tautomers.
  • the invention only provides the compound having the fragment Aa, the tautomer of the compound having the frament Ab is also been provided at the same time; though the invention only provides the compound having the fragment Ba, the tautomer of the compound having the frament Bb is also been provided at the same time.
  • compounds disclosed herein may optionally be substituted with one or more substituents, such as are illustrated generally below, or as exemplified by particular classes, subclasses, and species of the invention.
  • substituents such as are illustrated generally below, or as exemplified by particular classes, subclasses, and species of the invention.
  • the phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted” .
  • substituted refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent.
  • an optionally substituted group may have a substituent at each substitutable position of the group.
  • substituent When more than one position in a given structure can be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position.
  • substituent When a substituent is defined by using “independently selected from... ” means that the substituent is independent from each other, each selection of subsituent can be identical or different.
  • substituents of compounds disclosed herein are disclosed in groups or in ranges. It is specifically intended that the invention include each and every individual subcombination of the members of such groups and ranges.
  • C 1-6 alkyl is specifically intended to individually disclose methyl, ethyl, C 3 alkyl, C 4 alkyl, C 5 alkyl, and C 6 alkyl.
  • linking substituents are described. Where the structure clearly requires a linking group, the Markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the Markush group definition for that variable lists “alkyl” or “aryl” then it is understood that the “alkyl” or “aryl” represents a linking alkylene group or arylene group, respectively.
  • alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon group of 1-20 carbon atoms, wherein the alkyl group is optionally substituted with one or more substituents described herein. Unless otherwise stated, the alkyl group contains 1-20 carbon atoms. In some embodiments, the alkyl group contains 1-12 carbon atoms. In other embodiments, the alkyl group contains 1-6 carbon atoms. In still other embodiments, the alkyl group contains 1-4 carbon atoms. In yet other embodiments, the alkyl group contains 1-3 carbon atoms.
  • alkyl group examples include, methyl (Me, -CH 3 ) , ethyl (Et, -CH 2 CH 3 ) , n-propyl (n-Pr, -CH 2 CH 2 CH 3 ) , isopropyl (i-Pr, -CH (CH 3 ) 2 ) , n-butyl (n-Bu, -CH 2 CH 2 CH 2 CH 3 ) , isobutyl (i-Bu, -CH 2 CH (CH 3 ) 2 ) , sec-butyl (s-Bu, -CH (CH 3 ) CH 2 CH 3 ) , tert-butyl (t-Bu, -C (CH 3 ) 3 ) , n-pentyl (-CH 2 CH 2 CH 2 CH 3 ) , 2-pentyl (-CH (CH 3 ) CH 2 CH 2 CH 3 ) , 3-pentyl (-CH (CH 2 CH 3 )
  • haloalkyl or “haloalkoxy” refer to alkyl, or alkoxy, as the case may be, substituted with one or more halogen atoms.
  • haloalkyl or “haloalkoxy” groups include trifluoromethyl, trifluoromethoxy, and the like.
  • hydroxyalkyl or “hydroxy substituted alkyl” refers to an alkyl group substituted with one or more hydroxy groups, wherein the alkyl is as defined herein. Some non-limiting examples include hydroxymethyl, hydroxyethyl, 1, 2-dihydroxyethyl, and the like.
  • aminoalkyl or “amino substituted alkyl” refers to an alkyl group substituted with one or more amino groups, wherein the alkyl is as defined herein.
  • alkoxy refers to an alkyl group, as previously defined, attached to the parent molecular moiety via an oxygen atom. Unless otherwise specified, the alkoxy group contains 1-12 carbon atoms. In one embodiment, the alkoxy group contains 1-6 carbon atoms. In other embodiment, the alkoxy group contains 1-4 carbon atoms. In still other embodiment, the alkoxy group contains 1-3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents disclosed herein.
  • alkoxyalkyl refers to an alkoxy group attached to the rest of the molecule via an alkyl group, wherein the alkoxy and alkyl are as defined herein.
  • alkoxyalkyl include, but are not limited to, methoxymethyl, methoxyethyl, 2-methoxyethyl, methoxypropyl, ethoxymethyl, ethoxyethyl, 1-propoxymethyl, 2-propoxyethyl, 1-butoxymethyl, 2-methyl-1-propoxyethyl, 2-butoxymethyl, and the like.
  • alkylacyl or “alkoxyacyl” refers to an alkyl group or an alkoxy group, as previously defined, attached to the parent molecular moiety via acyl group, wherein the alkyl, alkoxy and acyl are as defined herein.
  • alkylacyloxy or “alkoxyacyloxy” refers to an alkyl group or an alkoxy group, as previously defined, attached to the parent molecular moiety via acyloxy group, wherein the alkyl, alkoxy and acyloxy are as defined herein.
  • M atom of acyl group is carbon and sulfur.
  • haloalkoxyalkyl refers to a haloalkoxy group attached to the rest of the molecule through an alkyl group, wherein the haloalkoxy and alkyl are as defined herein.
  • alkylamino refers to “N-alkylamino” and “N, N-dialkylamino” , wherein amino groups are independently substituted with one alkyl radical or two alkyl radicals, respectively.
  • the alkylamino group is a lower alkylamino group having one or two C 1-6 alkyl groups attached to nitrogen atom.
  • the alkylamino group is a lower alkylamino group having one or two C 1-3 alkyl groups attached to nitrogen atom.
  • alkylamino group examples include monoalkylamino or dialkylamino, such as N-methylamino, N-ethylamino, N, N-dimethylamino, N, N-diethylamino, and the like.
  • alkylaminoalkyl refers to an alkylamino group attached to the rest of the molecule through an alkyl group, wherein the alkylamino and alkyl are as defined herein.
  • alkylthio refers to an alkyl group, as previously defined, attached to the parent molecular moiety via a sulfur atom, wherein the alkyl is as defined herein.
  • alkylthioalkyl refers to an alkylthio group attached to the rest of the molecule via an alkyl group, wherein the alkylthio and alkyl are as defined herein.
  • alkenyl refers to a linear or branched-chain monovalent hydrocarbon radical of 2 to 12 carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp 2 double bond, wherein the alkenyl radical may be optionally substituted with one or more substituents described herein, and includes radicals having “cis” and “trans” orientations, or alternatively, “E” and “Z” orientations.
  • the alkenyl contains 2 to 8 carbon atoms.
  • the alkenyl contains 2 to 6 carbon atoms.
  • the alkenyl contains 2 to 4 carbon atoms.
  • alkynyl refers to a linear or branched monovalent hydrocarbon radical of 2 to 12 carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally substituted with one or more substituents described herein.
  • the alkynyl contains 2 to 8 carbon atoms.
  • the alkynyl contains 2 to 6 carbon atoms.
  • the alkynyl contains 2 to 4 carbon atoms. Examples of such groups include, but are not limited to, ethynyl (-C ⁇ CH) , propargyl (-CH 2 C ⁇ CH) , 1-propynyl (-C ⁇ C-CH 3 ) , and the like.
  • cycloalkyl refers to a monovalent or multivalent saturated ring having 3 to 12 carbon atoms as a monocyclic, bicyclic, or tricyclic ring system. In some embodiments, the cycloalkyl group contains 3 to 12 carbon atoms. In other embodiments, the cycloalkyl group contains 3 to 8 carbon atoms. In still other embodiments, the cycloalkyl group contains 3 to 6 carbon atoms.
  • cycloalkylalkyl refers to a cycloalkyl group attached to the rest of the molecule via an alkyl group, wherein the cycloalkyl and alkyl are as defined herein.
  • heterocyclyl and “heterocycle” as used interchangeably herein refer to a saturated or partially unsaturated monocyclic, bicyclic or tricyclic ring containing 3-12 ring atoms of which at least one ring member is selected from nitrogen, sulfur and oxygen and at least one ring is nonaromatic.
  • the sulfur can be optionally oxygenized to S-oxide and the nitrogen can be optionally oxygenized to N-oxide.
  • heterocyclyl group examples include oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiolanyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thioxanyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, o
  • heterocyclyl wherein the ring sulfur atom is oxidized is sulfolanyl, 1, 1-dioxo-thiomorpholinyl.
  • the heterocyclyl group may be optionally substituted with one or more substituents disclosed herein.
  • heterocyclyl may be 3-8 membered heterocyclyl, which refers to a saturated or partially unsaturated monocyclic ring containing 3-8 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen.
  • the sulfur can be optionally oxygenized to S-oxide and the nitrogen can be optionally oxygenized to N-oxide.
  • heterocyclyl group containing 4-7 ring atoms examples include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiolanyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thioxanyl, homopiperazinyl, homopiperidinyl, oxepanyl, thie
  • heterocyclyl wherein the ring sulfur atom is oxidized is sulfolanyl, 1, 1-dioxo-thiomorpholinyl.
  • the heterocyclyl group containing 3-8 ring atoms may be optionally substituted with one or more substituents disclosed herein.
  • heterocyclyl may be 4 membered heterocyclyl, which refers to a saturated or partially unsaturated monocyclic ring containing 4 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen.
  • the sulfur can be optionally oxygenized to S-oxide and the nitrogen can be optionally oxygenized to N-oxide.
  • heterocyclyl containing 4 ring atoms include azetidinyl, oxetanyl and thietanyl.
  • the heterocyclyl group containing 4 ring atoms may be optionally substituted with one or more substituents disclosed herein.
  • heterocyclyl may be 5 membered heterocyclyl, which refers to a saturated or partially unsaturated monocyclic ring containing 5 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen.
  • the sulfur can be optionally oxygenized to S-oxide and the nitrogen can be optionally oxygenized to N-oxide.
  • heterocyclyl examples include, but are not limited to, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiolanyl.
  • heterocyclyl wherein the ring sulfur atom is oxidized include sulfolanyl.
  • the heterocyclyl group containing 5 ring atoms may be optionally substituted with one or more substituents disclosed herein.
  • heterocyclyl may be 6 membered heterocyclyl, which refers to a saturated or partially unsaturated monocyclic ring containing 6 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen.
  • the sulfur can be optionally oxygenized to S-oxide and the nitrogen can be optionally oxygenized to N-oxide.
  • heterocyclyl group containing 6 ring atoms examples include, but are not limited to, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl and thioxanyl.
  • heterocyclyl wherein the ring sulfur atom is oxidized include 1, 1-dioxo-thiomorpholinyl.
  • the heterocyclyl group containing 6 ring atoms may be optionally substituted with one or more substituents disclosed herein.
  • heterocyclyl may be 7-12 membered heterocyclyl, which refers to a saturated or partially unsaturated spiro or fused bicyclyl ring containing 7-12 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen.
  • the sulfur can be optionally oxygenized to S-oxide and the nitrogen can be optionally oxygenized to N-oxide.
  • heterocyclyl containing 7-12 ring atoms include indolinyl, 1, 2, 3, 4-tetrahydroisoquinolyl, 1, 3-benzodioxolyl, 2-oxa-5-azabicyclo [2.2.1] hept-5-yl.
  • the heterocyclyl group containing 7-12 ring atoms may be optionally substituted with one or more substituents disclosed herein.
  • heterocyclyloxy or “heterocyclylamino” refers to a heterocyclyl group attached to the rest part of the molecule through oxygen atom or nitrogen atom. Wherein the heterocyclyl group is as defined herein.
  • heterocyclylalkyl refers to an alkyl group substitued with heterocyclyl.
  • heterocyclylalkoxy refers to an alkoxy group substitued with heterocyclyl, which attached to the rest of molecular through an oxygen atom.
  • heterocyclylalkylamino refers to an alkylamino group substitued with heterocyclyl, which attached to the rest of molecular through nitrogen atom.
  • Some non-limiting examples of such groups include pyrrol-2-yl-methyl, morpholin-4-yl-ethyl, morpholin-4-yl-ethoxy, piperazin-4-yl-ethoxy, piperidin-4-yl-ethylamino and the like.
  • aryl refers to monocyclic, bicyclic or tricyclic carbocyclic ring system having a total of six to fourteen ring members, or six to twelve ring members, or six to ten ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3 to 7 ring members and that has a single point or multipoint of attachment to the rest of the molecule.
  • aryl and “aromatic ring” can be used interchangeably herein. Examples of aryl group may include phenyl, naphthyl and anthracene. The aryl group may be optionally and independently substituted with one or more substituents disclosed herein.
  • aryloxy refers to an aryl group, attached to the rest part of the molecule through oxygen atom. Wherein the aryl group is as defined herein.
  • arylamino refers to an amino group substituted with one or two aryl groups, wherein the aryl is as defined herein. Some non-limiting examples of such group included N-phenylamino. In some embodiments, the aryl group of the arylamino may be further substituted.
  • arylalkyl refers to an aryl substituted alkyl group, which attached to the rest of the molecule via an alkyl group
  • arylalkoxy refers to an aryl substituted alkoxy group, which attached to the rest of the molecule via oxygen atom
  • arylalkylamino refers to an aryl substituted alkylamino group, which attached to the rest of the molecule via the nitrogen atom.
  • heteroaryl refers to monocyclic, bicyclic or tricyclic ring system having a total of five to twelve ring members, or five to ten ring members, or five to six ring members, wherein at least one ring in the system is aromatic, and in which at least one ring member is selected from heteroatom, and wherein each ring in the system contains 5 to 7 ring members and that has a single point or multipoint of attachment to the rest of the molecule.
  • heteroaryl and “heteroaromatic ring” or “heteroaromatic compound” can be used interchangeably herein.
  • the heteroaryl group is optionally substituted with one or more substituents disclosed herein.
  • a 5-10 membered heteroaryl comprises 1, 2, 3 or 4 heteroatoms independently selected from O, S and N.
  • heteroaryl rings include 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl) , 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl) , tri
  • heteroaryloxy refers to a heteroaryl group, attached to the rest part of the molecule through oxygen atom. Wherein the heteroaryl group is as defined herein.
  • heteroarylamino refers to an amino group substituted with one or two heteroaryl groups.
  • heteroarylalkyl refers to an heteroaryl substituted alkyl group, which attached to the rest of the molecule via an alkyl group
  • heteroarylalkoxy refers to an heteroaryl substituted alkoxy group, which attached to the rest of the molecule via the oxygen atom
  • heteroarylalkylamino refers to an heteroaryl substituted alkylamino group, which attached to the rest of the molecule via the nitrogen atom.
  • fused bicycle and “fused bicyclyl” as used interchangeably herein refer to a monovalent or multivalent saturated or partially unsaturated bridged ring system, which refers to a nonaromatic bicyclic ring system. Such system can comprises independent or conjugated unsaturated units, but the core structure does not comprise aromatic ring or heteroaromatic ring (but aromatic group can be used as substituents of the fused bicyclyl group) . In some embodiments, the fused bicyclyl group contains 5 to 12 carbon atoms.
  • spiro bicyclyl or “spiro bicyclic” as used interchangeably herein refers to a monovalent or multivalent saturated or partially unsaturated ring system, wherein a ring originats from a particular annular carbon of another ring.
  • the spiro bicyclyl group contains 5 to 12 carbon atoms.
  • a saturated bridged ring system (ring B and B’) is termed as “fused bicyclic” , whereas ring A and ring B share an atom between the two saturated ring system, which terms as a “spirocyclyl” or “spiro bicyclyl” .
  • fused heterobicyclyl or “spiro heterobicyclyl” , each ring of which may be carbon or nitrogen linked.
  • fused heterobicyclyl group or spiro heterobicyclyl group contains 5 to 12 carbon atoms.
  • hydroxy refers to -OH.
  • amino refers to -NH 2 .
  • cyano refers to -CN.
  • mercapto refers to -SH.
  • nitro refers to -NO 2 .
  • halogen refers to fluorine (F) , chlorine (Cl) , bromine (Br) or iodine (I) .
  • heteroatom refers to one or more of oxygen, sulfur, nitrogen, phosphorus and silicon, including any oxidized form of nitrogen, sulfur, or phosphorus; the quaternized form of any basic nitrogen; or a substitutable nitrogen of a heterocyclic ring, for example, N (as in 3, 4-dihydro-2H-pyrrolyl) , NH (as in pyrrolidinyl) or NR (as in N-substituted pyrrolidinyl) .
  • n membered where n is an integer typically describes the number of ring-forming atoms in a moiety where the number of ring-forming atoms is n.
  • piperidinyl is an example of a 6 membered heterocycloalkyl
  • 1, 2, 3, 4-tetrahydro-naphthalene is an example of a 10 membered carbocyclyl group.
  • unsaturated refers to a moiety having one or more units of unsaturation.
  • a bond drawn from a substituent to the center of one ring within a ring system represents substitution of the substituent (R 5 ) n , R 6 and (R 15 ) n1 at any substitutable position on the ring.
  • protecting group refers to a substituent that is commonly employed to block or protect a particular functionality while reacting with other functional groups on the compound.
  • an “amino-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxy-carbonyl (BOC, Boc) , benzyloxycarbonyl (CBZ, Cbz) and 9-fluorenylmethylenoxy-carbonyl (Fmoc) .
  • a “hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality.
  • Suitable protecting groups include acetyl and silyl.
  • a “carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality. Common carboxy-protecting groups include -CH 2 CH 2 SO 2 Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxy-methyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrophenylsulfonyl) ethyl, 2- (diphenylphosphino) ethyl, nitroethyl and the like.
  • protecting groups and their use see T.W. Greene, Protective Groups in Organic Synthesis, John Wiley &Sons, New York, 1991; and P.J. Kocienski, Protecting Groups, Thieme, Stuttgart, 2005.
  • prodrug refers to a compound that is transformed in vivo into a compound of Formula (I) , (II) , (IIa) , (IIb) and (III) . Such a transformation can be affected, for example, by hydrolysis of the prodrug form in blood or enzymatic transformation to the parent form in blood or tissue.
  • Prodrugs of the compounds disclosed herein may be, for example, esters. Some common esters which have been utilized as prodrugs are phenyl esters, aliphatic (C 1-24 ) esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters.
  • a compound disclosed herein that contains a hydroxy group may be acylated at this position in its prodrug form.
  • Other prodrug forms include phosphates, such as, those phosphate compounds derived from the phosphonation of a hydroxy group on the parent compound.
  • a “metabolite” is a product produced through metabolism in the body of a specified compound or salt thereof.
  • the metabolites of a compound may be identified using routine techniques known in the art and their activities determined using tests such as those described herein. Such products may result for example from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzyme cleavage, and the like, of the administered compound.
  • the invention includes metabolites of compounds disclosed herein, including metabolites produced by contacting a compound disclosed herein with a mammal for a sufficient time period.
  • a “pharmaceutically acceptable salts” refers to organic or inorganic salts of a compound disclosed herein.
  • Pharmaceutically acceptable salts are well known in the art. For example, S.M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66: 1-19, which is incorporated herein by reference.
  • Some non-limiting examples of pharmaceutically acceptable and nontoxic salts include salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid and malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid and malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, laurylsulfate, malate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil soluble or dispersable products may be obtained by such quaternization.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, C 1-8 sulfonate or aryl sulfonate.
  • solvate refers to an association or complex of one or more solvent molecules and a compound disclosed herein.
  • solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid and ethanolamine.
  • hydrate refers to the complex where the solvent molecule is water.
  • the term “treat” , “treating” or “treatment” of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof) .
  • “treat” , “treating” or “treatment” refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient.
  • “treat” , “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom) , physiologically, (e.g., stabilization of a physical parameter) , or both.
  • “treat” , “treating” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen
  • 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, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic 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, ammonium salts and metals from columns I to XII of the periodic table.
  • the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like.
  • Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from a basic or acidic moiety, by conventional chemical methods.
  • such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like) , or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid.
  • a stoichiometric amount of the appropriate base such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like
  • Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two.
  • use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable.
  • the compounds disclosed herein, including their salts can also be obtained in the form of their hydrates, or include other solvents such as ethanol, DMSO, and the like, used for their crystallization.
  • the compounds of the present invention may inherently or by design form solvates with pharmaceutically acceptable solvents (including water) ; therefore, it is intended that the invention embrace both solvated and unsolvated forms.
  • any formula given herein is also intended to represent isotopically unenriched forms as well as isotopically enriched forms of the compounds. Any formula given herein is also intended to represent isotopically unenriched forms as well as isotopically enriched forms of the compounds.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, and chlorine, such as 2 H (deuterium, D) , 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 18 F, 31 P, 32 P, 35 S, 36 Cl, 125 I, respectively.
  • the compounds of the invention include isotopically enriched compounds as defined herein, for example those into which radioactive isotopes, such as 3 H, 14 C and 18 F, or those into which non-radioactive isotopes, such as 2 H and 13 C are present.
  • isotopically enriched compounds are useful in metabolic studies (with 14 C) , reaction kinetic studies (with, for example 2 H or 3 H) , detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F-enriched compound may be particularly desirable for PET or SPECT studies.
  • Isotopically-enriched compounds of Formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a substituent in a compound of this invention is denoted deuterium
  • such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5%deuterium incorporation at each designated deuterium atom) , at least 4000 (60%deuterium incorporation) , at least 4500 (67.5%deuterium incorporation) , at least 5000 (75%deuterium incorporation) , at least 5500 (82.5%deuterium incorporation) , at least 6000 (90%deuterium incorporation) , at least 6333.3 (95%deuterium incorporation) , at least 6466.7 (97%deuterium incorporation) , at least 6600 (99%deuterium incorporation) , or at least 6633.3 (99.5%deuterium incorporation) .
  • Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D 2 O, acetone-d 6 , DMSO-d
  • provided herein is a compound for preparing the compound of Formula (I) , (II) , (IIa) , (IIb) and (III) .
  • provided herein is a method of preparing, separating or purifying the compound of Formula (I) , (II) , (IIa) , (IIb) and (III) .
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the compounds disclosed herein and pharmaceutically acceptable carrier, excipient, diluent, adjuvant, solvent or a combination thereof.
  • the pharmaceutical composition can be liquid, solid, semisolid, gel or spray.
  • ком ⁇ онент there is meant either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound of the invention and a combination partner may be administered independently at the same time or separately within time intervals that especially allow that the combination partners show a cooperative, e.g. synergistic effect.
  • co-administration or “combined administration” or the like as used herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
  • pharmaceutical combination refers to a product obtained from mixing or combining active ingredients, and includes both fixed and non-fixed combinations of the active ingredients.
  • fixed combination means that the active ingredients, e.g., a compound of the invention and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage.
  • non-fixed combination means that the active ingredients, e.g. a compound of the invention and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the active ingredients in the body of the patient.
  • cocktail therapy e.g., the administration of three or more active ingredients.
  • inhibiting HCV viral protein should be broadly understood, which comprises inhibiting the expression level of HCV viral protein, inhibiting activity level of HCV viral protein, viral assembly and egress level.
  • the expression level of HCV protein includes but not limited to, translation level of the viral protein, posttranslational modification level of the viral protein, replication level of genetic material in offsprings and so on.
  • the present invention refers to a method of anti HCV infection.
  • the compounds or composition thereof disclosed herein have good inhibitory effect on HCV infection.
  • a compound having Formula (I) or a stereoisomer a geometric isomer, a tautomer, an enantiomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof,
  • each of X and X 1 is independently N or CR 7b ;
  • each of R 1 , R 2 , R 3 and R 4 is independently H, deuterium, alkyl, arylalkyl, cycloalkyl, heterocyclyl, heteroaryl or aryl; or R 1 and R 2 , together with the X-CH to which they are attached, optionally form a 3-8 membered heterocyclic ring, 3-8 membered carbocyclic ring, fused bicyclic ring, fused heterobicyclic ring, spiro bicyclic ring or spiro heterobicyclic ring; or R 3 and R 4 , together with the X 1 -CH to which they are attached, optionally form a 3-8 membered heterocyclic ring, 3-8 membered carbocyclic ring, fused bicyclic ring, fused heterobicyclic ring, spiro bicyclic ring or spiro heterobicyclic ring;
  • each f, n and t is independently 0, 1, 2, 3 or 4;
  • n 1, 2, 3 or 4;
  • each r is independently 0, 1 or 2;
  • each alkyl, alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl, arylalkyl, -C ( O) - (CR 8 R 8a ) t -N (R 9 ) -R 10 , ⁇ -amino acid group, 3-8 membered heterocyclic ring, 3-8 membered carbocyclic ring, fused bicyclic ring, fused heterobicyclic ring, spiro bicyclic ring, spiro heterobicyclic ring, haloalkyl, alkenyl, alkynyl, alkoxyalkyl, heteroarylalkyl, cycloalkylalkyl, heterocyclylalkyl, arylamino, heteroarylamino, arylalkylamino, heteroarylalkylamino, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, heterocyclyloxy, heterocyclyl
  • A is -O-, -S-, -NH-, -CH 2 -S-, -CH 2 -O-, -CH 2 -NH-, -O-CH 2 -, -NH-CH 2 -, -S-CH 2 -or -CH 2 -CH 2 -.
  • each of R 1 , R 2 , R 3 and R 4 is independently H, C 1-6 alkyl, C 6-10 aryl-C 1-6 -alkyl, C 3-10 cycloalkyl, C 2-10 heterocyclyl, C 1-9 heteroaryl or C 6-10 aryl; or R 1 and R 2 , together with the X-CH to which they are attached, optionally form a 3-8 membered heterocyclic ring, 3-8 membered carbocyclic ring, C 5-12 fused bicyclic ring, C 5-12 fused heterobicyclic ring, C 5-12 spiro bicyclic ring or C 5-12 spiro heterobicyclic ring; or R 3 and R 4 , together with the X 1 -CH to which they are attached, optionally form a 3-8 membered heterocyclic ring, 3-8 membered carbocyclylic ring, C 5-12 fused bicyclic ring, C 5-12 fused heterobicyclic ring
  • R 1 and R 2 together with Y-X-CH to which they are attached, form one of the following groups:
  • R 3 and R 4 together with Y 1 -X 1 -CH to which they are attached, form one of the following groups:
  • each R 9b is independently H, deuterium, C 1-6 alkyl, C 1-6 haloalkyl, C 1-6 hydroxyalkyl, C 1-6 aminoalkyl, C 1-6 alkoxy-C 1-6 -alkyl, C 1-6 alkylamino-C 1-6 -alkyl, C 1-6 alkylthio-C 1-6 -alkyl, C 6-10 aryl-C 1-6 -alkyl, C 1-9 heteroaryl, C 6-10 aryl, C 2-10 heterocyclyl or C 3-8 cycloalkyl.
  • each R 9b is independently H, deuterium, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 hydroxyalkyl, C 1-3 aminoalkyl, C 1-3 alkoxy-C 1-3 -alkyl, C 1-3 alkylamino-C 1-3 -alkyl, C 1-3 alkylthio-C 1-3 -alkyl, C 6-10 aryl-C 1-3 -alkyl, C 1-9 heteroaryl, C 6-10 aryl, C 2-10 heterocyclyl or C 3-8 cycloalkyl.
  • each R 9b is independently H, deuterium, methyl, ethyl, propyl, isopropyl, trifluoromethyl, hydroxymethyl, aminomethyl, methoxymethyl, ethoxymethyl, phenylmethyl, phenyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
  • each r is independently 0, 1 or 2.
  • each r is independently 0, 1 or 2.
  • each R 8 and R 8a is independently H, deuterium, hydroxy, amino, F, Cl, Br, I, cyano, C 1-6 alkyl, C 1-6 deuterated alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 2-10 heterocyclyl, C 3-8 cycloalkyl, mercapto, nitro, C 6-10 aryl-C 1-6 -alkyl, C 1-6 alkoxy-C 1-6 -alkyl, C 1-9 heteroaryl-C 1-6 -alkyl, C 3-8 cycloalkyl-C 1-6 -alkyl, C 2-10 heterocyclyl-C 1-6 -alkyl, C 6-10 arylamino, C 1-9 heteroarylamino, C 6-10 aryl-C 1-6 -alkylamino, C 1-9 heteroaryl-C 1-6 -alkylamino, C 1-9 heteroaryloxy, C 6
  • each R 8 and R 8a is independently H, deuterium, hydroxy, amino, F, Cl, Br, I, cyano, C 1-4 alkyl, C 1-4 deuterated alkyl, C 1-4 haloalkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 2-8 heterocyclyl, C 3-8 cycloalkyl, mercapto, nitro, C 6-10 aryl-C 1-3 -alkyl, C 1-3 alkoxy-C 1-3 -alkyl, C 1-9 heteroaryl-C 1-3 -alkyl, C 3-8 cycloalkyl-C 1-3 -alkyl, C 2-8 heterocyclyl-C 1-3 -alkyl, C 6-10 arylamino, C 1-9 heteroarylamino, C 6-10 aryl-C 1-3 -alkylamino, C 1-9 heteroaryl-C 1-3 -alkylamino, C 1-9 heteroaryloxy, C 6
  • each R 8 and R 8a is independently H, deuterium, hydroxy, amino, F, Cl, Br, I, cyano, methyl, ethyl, isopropyl, n-butyl, i-butyl, s-butyl, t-butyl, deuterated methyl, deuterated ethyl, deuterated propyl, deuterated isopropoxy, methoxy, ethoxy, isopropoxy, methoxymethyl, 1-methoxyethyl, 2-methoxyethyl, 1-methoxypropyl, 2-methoxypropyl, 3-methoxypropyl, phenyl, pyranyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, trifluoromethyl, vinyl, allyl, ethynyl, morpholinyl, mercapto, nitro, phenylmethyl or phenylamin
  • t is 0, 1, 2, 3 or 4.
  • each r is independently 0, 1 or 2.
  • each n 1 and n 2 is independently 1, 2, 3 or 4.
  • the compound of the invention has Formula (II) , (IIa) , (IIb) or (III) , or a stereoisomer, a geometric isomer, a tautomer, an enantiomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof,
  • the compound of the invention has one of the following structures, or a stereoisomer, a geometric isomer, a tautomer, an enantiomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof:
  • the compound of the invention (comprising a compound having Formula (I) , (II) , (IIa) , (IIb) or (III) , or a stereoisomer, a geometric isomer, a tautomer, an enantiomer, an N-oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof) can be used in the manufacture of a medicament for treatment of acute or chronic HCV infection including that described herein. Further more, the compound of the invention can be used in the manufacture of an anti-HCV medicament. The compound disclosed herein also can be used in the manufacture of a medicament for lessening,preventing, managing or treating diseases mediated by HCV.
  • the compound of the invention can act as an active ingredient of a pharmaceutical composition
  • the pharmaceutical composition may comprises the compound of Formula (I) , (II) , (IIa) , (IIb) or (III) , and may further comprises at least one pharmaceutically acceptable carrier, adjuvant or diluent.
  • the salt is a pharmaceutically acceptable salt.
  • pharmaceutically acceptable refers to that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • the skills in the art could choose “pharmaceutically acceptable” substance or composition based on the other ingredients and the objects for treatment such as human.
  • the compounds disclosed herein also include salts of the compounds which are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds of Formula (I) , (II) , (IIa) , (IIb) or (III) , and/or for separating enantiomers of compounds of Formula (I) , (II) , (IIa) , (IIb) or (III) .
  • the desired salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid or organic acid, wherein the inorganic acid comprises hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • organic acid examples include acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid and salicylic acid; a pyranosidyl acid, such as glucuronic acid and galacturonic acid; an alpha-hydroxy acid, such as citric acid and tartaric acid; an amino acid, such as aspartic acid and glutamic acid; an aromatic acid, such as benzoic acid and cinnamic acid; a sulfonic acid, such as p-toluenesulfonic acid, ethanesulfonic acid, and the like.
  • the desired salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary) , an alkali metal hydroxide, or alkaline earth metal hydroxide, and the like.
  • suitable salts include organic salts derived from amino acids, such as glycine and arginine; ammonia, such as primary, secondary and tertiary amine, and cyclic amines, such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, lithium, and the like.
  • the pharmaceutical composition disclosed herein comprises any one of the compounds of the invention.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle or a combination thereof.
  • the pharmaceutical composition can be used for treating HCV infection or a HCV disorder.
  • the pharmaceutical composition disclosed herein further comprises anti-HCV agents.
  • the anti-HCV agent may be any other known anti-HCV agent except the compound described herein.
  • the interferon is interferon ⁇ -2b, pegylated interferon ⁇ , interferon ⁇ -2a, pegylated interferon ⁇ -2a, consensus interferon- ⁇ , interferon ⁇ or a combination thereof.
  • the pharmaceutical composition futher comprise an HCV inhibitor, wherein the HCV inhibitor inhibits HCV replication process and/or at least a function of a HCV viral protein, and wherein the HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress; and wherein the HCV viral proteins comprise metalloproteinase, non-structural protein NS2, NS3, NS4A, NS4B, NS5A or NS5B, an internal ribosome entry site (IRES) and inosine-5’-monophosphate dehydrogenase (IMPDH) required in HCV viral replication.
  • HCV inhibitor inhibits HCV replication process and/or at least a function of a HCV viral protein
  • the HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress
  • the HCV viral proteins comprise metalloproteinase, non-structural protein NS2, NS3, NS4A,
  • compositions which comprise therapeutically effective amounts of compounds of Formula (I) , (II) , (IIa) , (IIb) or (III) , or pharmaceutically acceptable salts thereof, and one or more pharmaceutically acceptable carrier, diluent or excipient.
  • terapéuticaally effective amount refers to the total amount of each active component that is sufficient to show a meaningful patient benefit (e.g., a reduction in viral load) .
  • a meaningful patient benefit e.g., a reduction in viral load
  • the term refers to that ingredient alone.
  • the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially, or simultaneously.
  • the compounds of Formula (I) , (II) , (IIa) , (IIb) or (III) , and pharmaceutically acceptable salts thereof, are as described above.
  • the carrier (s) , diluent (s) , or excipient (s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • a process for the preparation of a pharmaceutical formulation including admixing a compound of Formula (I) , (II) , (IIa) , (IIb) or (III) , or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents, or excipients.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
  • compositions may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. Dosage levels of between about 0.01 and about 250 milligram per kilogram ( “mg/kg” ) body weight per day, preferably between about 0.05 and about 100 mg/kg body weight per day of the compounds of the present disclosure are typical in a monotherapy for the prevention and treatment of HCV mediated disease. Typically, the pharmaceutical compositions of this disclosure will be administered from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
  • mg/kg milligram per kilogram
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending on the condition being treated, the severity of the condition, the time of administration, the route of administration, the rate of excretion of the compound employed, the duration of treatment, and the age, gender, weight, and condition of the patient.
  • Preferred unit dosage formulations are those containing a daily dose or sub-dose, as herein above recited, or an appropriate fraction thereof, of an active ingredient. Treatment may be initiated with small dosages substantially less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached.
  • the compound is most desirably administered at a concentration level that will generally afford antivirally effective results without causing any harmful or deleterious side effects.
  • compositions of this disclosure comprise a combination of a compound of the present disclosure and one or more additional therapeutic or prophylactic agent
  • both the compound and the additional agent are usually present at dosage levels of between about 10 to 150%, and more preferably between about 10 and 80%of the dosage normally administered in a monotherapy regimen.
  • Pharmaceutical formulations may be adapted for administration by any appropriate route, for example by the oral (including buccal or sublingual) , rectal, nasal, topical (including buccal, sublingual, or transdermal) , vaginal, or parenteral (including subcutaneous, intracutaneous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional, intravenous, or intradermal injections or infusions) route.
  • Such formulations may be prepared by any method known in the art of pharmacy, for example by bringing into association the active ingredient with the carrier (s) or excipient (s) . Oral administration and administration by injection are preferred.
  • compositions adapted for oral administration may be presented as discrete units such as capsules or tablets; powders or granules; solution or suspensions in aqueous or non-aqueous liquids; edible foams or whips; or oil-in-water liquid emulsions or water-in-oil emulsions.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
  • an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like.
  • Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutical carrier such as an edible carbohydrate, as, for example, starch or mannitol. Flavoring, preservative, dispersing, and coloring agent can also be present.
  • Capsules are maded by preparing a powder mixture, as described above, and filling formed gelatin sheaths.
  • Glidants and lubricants such as colloidal silica, talc, magnesium stearate, calcium stearate, or solid polyethylene glycol can be added to the powder mixture before the filling operation.
  • a disintegrating or solubilizing agent such as agar-agar, calcium carbonate, or sodium carbonate can also be added to improve the availability of the medicament when the capsule is ingested.
  • suitable binders include starch, gelatin, natural sugars such as glucose or ⁇ -lactose, corn sweetener, natural and synthetic resin such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium chloride, and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, betonite, xanthan gum, and the like.
  • Tablets are formulated, for example, by preparing a powder mixture, granulating or slugging, adding a lubricant and disintegrant, and pressing into tablets.
  • a powder mixture is prepared by mixing the compound, suitable comminuted, with a diluents or base as described above, and optionally, with a binder such as carboxymethylcellulose, an alginate, gelating, or polyvinyl pyrrolidone, a solution retardant such as paraffin, a resorption accelerator such as a quaternary salt and/or and absorption agent such as betonite, kaolin, or dicalcium phosphate.
  • a binder such as carboxymethylcellulose, an alginate, gelating, or polyvinyl pyrrolidone
  • a solution retardant such as paraffin
  • a resorption accelerator such as a quaternary salt and/or
  • absorption agent such as betonite, kaolin, or dicalcium phosphate.
  • the powder mixture can be granulated by wetting with a binder such as syrup, starch paste, acadia mucilage, or solution of cellulosic or polymeric materials and forcing through a screen.
  • a binder such as syrup, starch paste, acadia mucilage, or solution of cellulosic or polymeric materials and forcing through a screen.
  • the powder mixture can be run through the tablet machine and the result is imperfectly formed slugs broken into granules.
  • the granules can be lubricated to prevent sticking to the tablet forming dies by means of the addition of stearic acid, a stearate salt, talc, or mineral oil.
  • the lubricated mixture is then compressed into tablets.
  • the compounds of the present disclosure can also be combined with a free flowing inert carrier and compressed into tablets directly without going through the granulating or slugging steps.
  • a clear or opaque protective coating consisting of a sealing coat of shellac,
  • Oral fluids such as solution, syrups, and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of the compound.
  • Syrups can be prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic vehicle.
  • Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxyethylene sorbitol ethers, preservatives, flavor additive such as peppermint oil or natural sweeteners, or saccharin or other artificial sweeteners, and the like can also be added.
  • dosage unit formulations for oral administration can be microencapsulated.
  • the formulation can also be prepared to prolong or sustain the release as for example by coating of embedding particulate material in polymers, wax, or the like.
  • the compounds of formula (I) , (II) , (IIa) , (IIb) or (III) , and pharmaceutically acceptable salts thereof may also be also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • the compounds of formula (I) , (II) , (IIa) , (IIb) or (III) , and pharmaceutically acceptable salts thereof may also be delivered by the use of monoclonal antibodies as individual carrier to which the compound molecules are coupled.
  • the compounds may also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers may encompass polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidophenol, polyhydroxyethylaspartamidophenol or polyethylene oxide polylysine, substituted by palmitoyl radicals.
  • the compounds may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, poly ( ⁇ -caprolactone) , polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross-linked or amphipathic block copolymers of hydrogels.
  • a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, poly ( ⁇ -caprolactone) , polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross-linked or amphipathic block copolymers of hydrogels.
  • compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmacol. Res., 1986, 3 (6) , 318.
  • compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols, oils or transdermal patch.
  • compositions adapted for rectal administration may be presented as suppositories or as enemas.
  • compositions adapted for nasal administration wherein the carrier is a solid include a course powder having a particle size for example in the range 20 to 500 microns which is administered in the manner in which snuff is taken, i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose.
  • suitable formulations wherein the carrier is a liquid, for administration as a nasal spray or nasal drops, include aqueous or oil solutions of the active ingredient.
  • Fine particle dusts or mists which may be generated by means of various types of metered, dose pressurized aerosols, nebulizers or insufflators or other devise which suitable for delivering aerosol spray.
  • compositions adapted for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulations.
  • compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain antioxidants, buffers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets.
  • formulations may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
  • the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for inhibiting HCV replication process and/or at least a function of a HCV viral protein
  • the HCV replication process is a whole viral cycle consisting of HCV entry, uncoating, translation, replication, assembly and egress
  • the HCV viral proteins comprise metalloproteinase, non-structural protein NS2, NS3, NS4A, NS4B, NS5A or NS5B, an internal ribosome entry site (IRES) and inosine-5’-monophosphate dehydrogenase (IMPDH) required in HCV viral replication.
  • any one of the compounds or the pharmaceutical compositions of the invention can be used for treating HCV infection or a HCV disorder.
  • a method which comprises administering the compound or the pharmaceutical composition disclosed herein, further comprising administering to the patient additional anti-HCV agents.
  • the compounds or the pharmaceutical compositions disclosed herein can be combined with additional anti-HCV agents.
  • the anti-HCV agent is an interferon, ribavirin, IL-2, IL-6, IL-12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, imiquimod, an inosine 5’-monophosphate dehydrogenase inhibitor, amantadine, rimantadine, bavituximab, hepatitis C immunogloblin, Civacir TM , boceprevir, telaprevir, erlotinib, daclatasvir, simeprevir, asunaprevir, vaniprevir, faldaprevir, paritaprevir, danoprevir, sovaprevir, grazoprevir
  • the treatment method that includes administering a compound or composition disclosed herein can further include administering to the patient an additional anti-HCV agent, wherein the additional anti-HCV drug is administered together with a compound or composition disclosed herein as a single dosage form or separately from the compound or composition as part of a multiple dosage form.
  • the additional anti-HCV agent may be administered at the same time as a compound disclosed herein or at a different time. In the latter case, administration may be staggered by, for example, 6 hours, 12 hours, 1 day, 2 days, 3 days, 1 week, 2 weeks, 3 weeks, 1 month, or 2 months.
  • an “effective amount” or “effective dose” of the compound or pharmaceutically acceptable composition is an amount that is effective in treating or lessening the severity of one or more of the aforementioned disorders.
  • the compounds and compositions, according to the method disclosed herein, may be administered using any amount and any route of administration which is effective for treating or lessening the severity of the disorder or disease. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • a compound or composition can also be administered with one or more other therapeutic agents as discussed above.
  • the compounds disclosed herein may be prepared by methods described herein, wherein the substituents are as defined for Formula (I) , (II) , (IIa) , (IIb) or (III) above, except where further noted.
  • the following non-limiting schemes and examples are presented to further exemplify the invention.
  • Anhydrous THF, dioxane, toluene, and ether were obtained by refluxing the solvent with sodium.
  • Anhydrous CH 2 Cl 2 and CHCl 3 were obtained by refluxing the solvent with CaH 2 .
  • EtOAc, PE, hexane, N, N-dimethylacetamide and DMF were treated with anhydrous Na 2 SO 4 prior to use.
  • reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried.
  • MS data were determined by an Agilent 6320 Series LC-MS spectrometer equipped with a G1312A binary pump and a G1316A TCC (column was operated at 30 °C) .
  • G1329A autosampler and G1315B DAD detector were applied in the analysis, and an ESI source was used in the LC-MS spectrometer.
  • MS data were determined by an Agilent 6120 Series LC-MS spectrometer equipped with a G1311A quaternary pump and a G1316A TCC (column was operated at 30 °C) .
  • G1329A autosampler and G1315D DAD detector were applied in the analysis, and an ESI source was used on the LC-MS spectrometer.
  • Compound s-10 can be prepared by a general synthetic procedure illustrated in Scheme 1, and wherein R 15 and R 8a are as defined herein.
  • Compound s-1 can react with trifluoromethanesulfonic anhydride under a base condition to afford compound s-2; compound s-2 can be bromized to afford compound s-3; compound s-3 can react with compound s-4 under a base condition to afford compound s-5; cyclization reaction of compound s-5 and ammonium acetate can afford compound s-6; compound s-6 can be oxidized to afford compound s-7; deprotection reaction of compound s-7 can afford compound s-8; condensation reaction of compound s-8 and compound s-9 can afford compound s-10.
  • Compound s-23 can be prepared by a general synthetic procedure illustrated in Scheme 2, and wherein R 15 and R 8 are as defined herein.
  • Compound s-11 can convert to compound s-12 in the present of a reductant; compound s-12 can be acetylized to afford compound s-13; demethylation reaction of compound s-13 can afford compound s-14; compound s-14 can react with trifluoromethanesulfonic anhydride under a base condtion to afford compound s-15; compound s-15 can be bromized to afford compound s-16, compound s-16 can react with compound s-17 under a base condition to afford compound s-18, compound s-18 can react with ammonium acetate to afford compound s-19; compound s-19 can convert to compound s-20 in the present of a Pd catalyst; deprotection reaction of compound s-20 can afford compound s-21; compound s-21 can react with s-22 to afford s-23.
  • Compound s-27 can be prepared by a general synthetic procedure illustrated in Scheme 3, and wherein R 15 and R 8 are as defined herein. Coupling reaction of compound s-8 and compound s-20 in the present of a Pd catalyst can afford compound s-24; deprotecting reaction of compound s-24 can afford compound s-25; condensation reaction of compound s-25 and compound s-26 can afford compound s-27.
  • Compound s-28 can be prepared by a general synthetic procedure illustrated in Scheme 4, and wherein R 15 , R 8 and R 8a are as defined herein. Coupling reaction of compound s-20 and compound s-23 in the present of a Pd catalyst can afford compound s-28.
  • HCV GT1a, GT1b and GT2a wild type replicons HCV GT3a, GT4a, GT5a and GT6a replicon chimeras
  • GT1b L31V, GT1b Y93H resistant strains HCV GT1a, GT1b and GT2a wild type replicons, HCV GT3a, GT4a, GT5a and GT6a replicon chimeras, and GT1b L31V, GT1b Y93H resistant strains.
  • Detection of activity of GT1a, GT1b and GT2a replicons HCV GT 1a H77 replicon, GT1b Con1b replicon and GT2a JFH1 replicon containing G418 resistance gene NEO and luciferase reporter gene were transfected respectively and immediately to Huh-7 cell by using an electric shock method. After the addition of G418, the cells were screened for 3 to 4 weeks, stable and transfected cell line was established. Cell line Huh7-H77 and Huh7-JFH1 was diluted to 5 ⁇ 10 4 /mL, 200 ⁇ L of which was seeded to a 96 well plate.
  • Cell line Huh7-Con1b was diluted to 1 ⁇ 10 5 /mL, 50 ⁇ L of which was seeded to a 384 well plate. 16-24 h later, the compound was diluted by 3-fold serially for a total of 11 concentrations to a suitable concentration, the diluted compound was added into the 96 well plate with POD TM 810 plate assembler, the final concentration of DMSO in each well was 0.5%.
  • the plate was incubated in a constant temperature incubator at 37 °C under 5%CO 2 for 72 h, and then to each well was added 40 ⁇ L of luciferase assay reagent (Promega Bright-Glo) , 5 min later, the plate was detected by chemiluminescence detection system (Envision) .
  • the results were processed by using GraphPad Prism software, EC50 of the compound against HCV relipcon was calculated.
  • the compound in DMSO mother solution was diluted, and the diluted solution was added to the 96 well plate, the final concentration of DMSO was 0.5%.
  • the cells were incubated under 5%CO 2 at 37 °C for 72 hours.
  • Luciferase luminescence substrate Bright-Glo was added to each well, 5 min later, the signal value of luminescence was detected by chemiluminescence detection system (Envision) , original data (RLU) were used for calculating inhibitory activity of the compound.
  • the corresponding curve of the compound were plotted and the value of the inhibitory activity thereof (EC50) against HCV relipcon were calculated by importing percent inhibition to GraphPad Prism software and nonlinear fitting.
  • the EC50 values of the compound against each HCV genotype replicon were listed in table 2.
  • One group was administered with test compound by intravenous injection at a dose of 1.0 mg/kg, and the other group was administered with test compound by oral gavage at a dose of 5.0 mg/kg.
  • Blood samples were collected at 8 to 9 time points within 24 hours after administration, and the standard curve was established based on the concentrations of the samples in a suitable range; the concentration of test compound in plasma sample was detected by using AB SCIEX API4000 LC-MS/MS in a MRM mode.
  • Pharmacokinetic parameters were calculated according to drug concentration -time curve using a noncompartmental method by WinNonLin 6.3 software.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Virology (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des composés en tant qu'inhibiteurs du virus de l'hépatite C et leurs utilisations pharmaceutiques. L'invention concerne spécifiquement des composés de formule (I) ou un stéréo-isomère, un tautomère, un énantiomère, un N-oxyde, un hydrate, un solvate, un métabolite, un sel pharmaceutiquement acceptable ou un promédicament de ceux-ci, qui peuvent être utilisés pour le traitement d'une infection par le virus de l'hépatite C (HVC) ou de troubles liés à l'hépatite C. En outre, l'invention concerne des compositions pharmaceutiques contenant le composé décrit ici et les procédés d'utilisation du composé ou des compositions pharmaceutiques les contenant dans le traitement d'une infection par le HVC ou des troubles du liés au HVC.
PCT/CN2016/076131 2015-03-12 2016-03-11 Composés en tant qu'inhibiteurs du virus de l'hépatite c et utilisations pharmaceutiques associées WO2016141890A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201510108484.6 2015-03-12
CN201510108484 2015-03-12
CN201510422786 2015-07-18
CN201510422786.0 2015-07-18

Publications (1)

Publication Number Publication Date
WO2016141890A1 true WO2016141890A1 (fr) 2016-09-15

Family

ID=56878945

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/076131 WO2016141890A1 (fr) 2015-03-12 2016-03-11 Composés en tant qu'inhibiteurs du virus de l'hépatite c et utilisations pharmaceutiques associées

Country Status (3)

Country Link
CN (1) CN105968101B (fr)
TW (1) TWI703141B (fr)
WO (1) WO2016141890A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10675358B2 (en) 2016-07-07 2020-06-09 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
WO2021197880A1 (fr) 2020-03-31 2021-10-07 Basf Se Procédé de préparation d'isoxazolines optiquement enrichies
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10004719B1 (en) * 2017-05-30 2018-06-26 Taigen Biotechnology Co., Ltd. Solid dispersion formulation
WO2024017234A1 (fr) * 2022-07-19 2024-01-25 广东东阳光药业股份有限公司 Sel d'inhibiteur de vhc, forme cristalline du sel, composition pharmaceutique du sel et son utilisation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102143959A (zh) * 2008-02-13 2011-08-03 百时美施贵宝公司 丙型肝炎病毒抑制剂
CN102741242A (zh) * 2009-12-16 2012-10-17 百时美施贵宝公司 丙型肝炎病毒抑制剂
CN104302290A (zh) * 2012-01-13 2015-01-21 百时美施贵宝公司 丙型肝炎病毒抑制剂

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190012271A (ko) * 2010-11-17 2019-02-08 길리애드 파마셋 엘엘씨 항바이러스 화합물
TWI610916B (zh) * 2012-08-03 2018-01-11 廣東東陽光藥業有限公司 作爲丙型肝炎抑制劑的橋環化合物及其在藥物中的應用

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102143959A (zh) * 2008-02-13 2011-08-03 百时美施贵宝公司 丙型肝炎病毒抑制剂
CN102741242A (zh) * 2009-12-16 2012-10-17 百时美施贵宝公司 丙型肝炎病毒抑制剂
CN104302290A (zh) * 2012-01-13 2015-01-21 百时美施贵宝公司 丙型肝炎病毒抑制剂

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10675358B2 (en) 2016-07-07 2020-06-09 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
US11110178B2 (en) 2016-07-07 2021-09-07 The Board Of Trustees Of The Leland Standford Junior University Antibody adjuvant conjugates
US11547761B1 (en) 2016-07-07 2023-01-10 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2
WO2021197880A1 (fr) 2020-03-31 2021-10-07 Basf Se Procédé de préparation d'isoxazolines optiquement enrichies

Also Published As

Publication number Publication date
CN105968101B (zh) 2019-03-01
CN105968101A (zh) 2016-09-28
TW201632514A (zh) 2016-09-16
TWI703141B (zh) 2020-09-01

Similar Documents

Publication Publication Date Title
CN108299532B (zh) 一种抗病毒核苷类似物前药及其组合物、用途
US9738629B2 (en) Bridged ring compounds as Hepatitis C virus inhibitors, pharmaceutical compositions and uses thereof
AU2018351400B2 (en) Dihydropyrimidine compounds and uses thereof in medicine
WO2015197028A1 (fr) Composés en tant qu'inhibiteurs du virus de l'hépatite c (vhc) et leurs utilisations en médecine
US9303061B2 (en) Spiro compounds as Hepatitis C virus inhibitors
US9309231B2 (en) Bridged ring compounds as hepatitis C virus (HCV) inhibitors and pharmaceutical applications thereof
US9504690B2 (en) Tetracyclic xanthene derivatives and methods of use thereof for the treatment of viral diseases
JP2021046456A (ja) Toll様レセプター調節4,6−ジアミノ−ピリド[3,2−d]ピリミジン化合物
WO2016015604A1 (fr) Composés en tant que petites molécules inhibitrices de la cdk et leurs utilisations
WO2018041091A1 (fr) Inhibiteurs de réplication du virus de la grippe, procédés d'application et utilisations associées
WO2016141890A1 (fr) Composés en tant qu'inhibiteurs du virus de l'hépatite c et utilisations pharmaceutiques associées
US9416139B2 (en) Spiro ring compound as hepatitis C virus (HCV) inhibitor and uses thereof
WO2016015597A1 (fr) Composés utilisés comme petits molécules inhibitrices de cdk et leurs utilisations
WO2016127859A1 (fr) Composés utilisés en tant qu'inhibiteurs du virus de l'hépatite c et leurs utilisations en médecine
AU2014352404A1 (en) Dihydropyrimidine compounds and their application in pharmaceuticals
WO2014148949A1 (fr) Alkyl 2-{[(2r,3s,5r)-5-(4-amino-2-oxo-2n-pyrimidin-1-yl)-3-hydroxy-tétrahydro-furan-2-ilméthoxy]-phénoxy-phosphorilamino}-propionates, inhibiteurs de nucléosides d'arn-polymérase hcv ns5b, procédés de fabrication et d'utilisation
US20150274739A1 (en) 2'-cyano substituted nucleoside derivatives and methods of use thereof for the treatment of viral diseases
CA3005921A1 (fr) Derives de (pyrrolo[2,3-b]pyridine-3-yl)-pyrimidine substituee et utilisation comme inhibiteurs de la replication du virus de la grippe
MX2013001088A (es) Derivados heterobiciclicos como inhibidores del virus de la hepatitis c.
WO2013059278A2 (fr) Inhibiteurs du virus de l'hépatite c
WO2014082381A1 (fr) Composés de type noyau spiro utilisables en tant qu'inhibiteurs du virus de l'hépatite c, compositions pharmaceutiques en contenant et leurs utilisations
WO2016095814A1 (fr) Composés cycliques pontés comme inhibiteurs du virus de l'hépatite c et préparation les comprenant
US20150232509A1 (en) Spiro ring compounds as hepatitis c virus (hcv) inhibitors
US9802949B2 (en) Fused ring compounds as hepatitis C virus inhibitors, pharmaceutical compositions and uses thereof
WO2014131315A1 (fr) Composés de type cycles à ponts à titre d'inhibiteurs du virus de l'hépatite c, compositions pharmaceutiques et leurs utilisations

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: 16761117

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 09/02/2018)

122 Ep: pct application non-entry in european phase

Ref document number: 16761117

Country of ref document: EP

Kind code of ref document: A1