WO2007056120A1 - Hepatitis c virus inhibitors - Google Patents

Hepatitis c virus inhibitors Download PDF

Info

Publication number
WO2007056120A1
WO2007056120A1 PCT/US2006/042922 US2006042922W WO2007056120A1 WO 2007056120 A1 WO2007056120 A1 WO 2007056120A1 US 2006042922 W US2006042922 W US 2006042922W WO 2007056120 A1 WO2007056120 A1 WO 2007056120A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
hcv
mmol
compound
halo
Prior art date
Application number
PCT/US2006/042922
Other languages
French (fr)
Inventor
Stanley D'andrea
Zhizhen Barbara Zheng
Jeffrey Allen Campbell
Fiona Mcphee
Andrew Charles Good
Paul Michael Scola
Original Assignee
Bristol-Myers Squibb Company
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 Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to EP06836865.3A priority Critical patent/EP1951743B1/en
Publication of WO2007056120A1 publication Critical patent/WO2007056120A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/204IL-6
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/208IL-12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • 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
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06139Dipeptides with the first amino acid being heterocyclic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic

Definitions

  • the present disclosure is generally directed to antiviral compounds, and more specifically directed to compounds which inhibit the functioning of the NS 3 protease (also referred to herein as "serine protease”) encoded by Hepatitis C virus (HCV) 3 compositions comprising such compounds and methods for inhibiting the functioning of the NS3 protease.
  • NS 3 protease also referred to herein as "serine protease”
  • 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. (Lauer, G. M.; Walker, B. D. N. Engl. J. Med. (2001), 345, 41-52).
  • 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.
  • Considerable heterogeneity is found within the nucleotide and encoded amino acid sequence throughout the HCV genome. At least six major genotypes have been characterized, and more than 50 subtypes have been described. The major genotypes of HCV differ in their distribution worldwide, and the clinical significance of the genetic heterogeneity of HCV remains elusive despite numerous studies of the possible effect of genotypes on pathogenesis and therapy.
  • 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 cleave at the NS2-NS3 junction; the second one is a serine protease contained within the N-terminal region of NS3 and mediates all the subsequent cleavages downstream of NS3, both in cis, at the NS3-NS4A cleavage site, and in trans, for the remaining NS4A- NS4B, NS4B-NS5A, NS5 A-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.
  • NS3 protein The complex formation of the NS3 protein with NS4A seems necessary to the processing events, enhancing the proteolytic efficiency at all of the sites.
  • the NS3 protein also exhibits nucleoside triphosphatase and RNA helicase activities.
  • NS5B is a RNA-dependent RNA polymerase that is involved in the replication of HCV.
  • R »4 is H; Ci -6 alkyl, or C 3-7 cycloalkyl each optionally substituted with halo; alkoxy; -C(O)-R 5 ; C(O)-N(R 5 ) 2 ; or C(O)-OR 5 ; wherein each R 5 is independently Ci- 9 alkyl, optionally substituted with C 1-6 alkoxy, C 3-7 cycloalkoxy, halo-Ci- 6 alkoxy, cyano, halo, hydroxy, amino, Ci -6 alkylamino, di (Ci- 6 ) alkylamino, di (Ci- 6 ) alkylamide, carboxyl, (Ci - ⁇ ) carboxyester; Ce-io aryl; C 7-14 alkylaryl; heterocylclyl; or C 3-7 cycloalkyl optionally substituted with alkoxy, halo, or haloalkoxy;
  • R 6 is H, C 1-6 alkyl, or C 3-7 cycloalkyl
  • R 3 and R 3 ' are each independently H or methyl;
  • Q is a C 3 . 9 saturated or unsaturated chain, optionally containing one to three heteroatoms independently selected from O or S(O) m ; wherein m is 0, 1 or 2;
  • W is OH, -O-R 1 , or -NH-SO 2 -R 2 ; wherein R 1 is d. 6 alkyl, unsaturated C 3-7 cycloalkyl, C 6 - I4 aryl or C 7-16 alkylaryl; and R 2 is Ci- 8 alkyl, C 4- I 0 alkylcycloalkyl, unsubstituted C 3-7 cycloalkyl; or R 2 is cyclopropyl or cyclobutyl optionally substituted with C 1 ⁇ alkyl, C 2-5 alkenyl, C 7 .
  • X is O, S, SO, SO 2 , OCH 2 , CH 2 O or NH;
  • R' is Het, C 6 -Io aryl or C 7 -I 4 alkylaryl, each optionally substituted with one or more R a groups; or R' is C 3-9 cycloalkyl or Ci- 7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; provided that when X is O, then R' is other than
  • R a> is H or R a ;
  • R x is selected from H, halo, Ci_ 6 alkyl, Ci -6 alkoxy, C 3-6 cycloalkyloxy, aryl, and Het; and
  • R y is selected from H, halo, CF 3 , Ci -6 alkoxy, and C 3-6 cycloalkyloxy; and (h) each R a is Ci- ⁇ alkyl, C3-7 cycloalkyl, CL 6 alkoxy, C 3-7 cycloalkoxy, halo-Ci- 6 alkyl, CF 3 , mono-or di-halo-Ci.
  • the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein X is O.
  • the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein X is O and R' is other than quinoline or isoquinoline.
  • the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein X is selected from S, SO, SO 2 , OCH 2 , CH 2 O and NH.
  • X is selected from S, SO, SO 2 , OCH 2 , CH 2 O and NH.
  • the present disclosure provides a compound of formula (II)
  • R 4 is -C(O)-R 5 , C(O)-NHR 5 or C(O)-OR 5 ; wherein R 5 is Ci -9 alkyl optionally substituted with C 1 . 6 alkoxy, cyano, halo;
  • Q is a C5- 7 saturated or unsaturated chain optionally containing one or two heteroatoms independently selected from O and S(O) m ; wherein m is 0, 1 or 2;
  • W is OH or NH-SO 2 -R 2 , wherein R 2 is Ci -8 alkyl, C 4-I0 alkylcycloalkyl, unsubstituted C 3 . 7 cycloalkyl, or cyclopropyl or cyclobutyl optionally substituted with Ci -4 alkyl or C 7 -Ig alkylaryl;
  • X is O, S, OCH 2 , CH 2 O or NH;
  • R' is Het, C 6-I0 aryl or C 7 - M alkylaryl, each optionally substituted with one or more R a ; or R' is C 3 . 9 cycloalkyl or Ci -7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; provided that when X is O, then R' is other than
  • R a' is H or R a ;
  • R x is selected from H, halo, C 1 - ⁇ alkyl, Cj -6 alkoxy, C 3-6 cycloalkyloxy, aryl, and Het; and
  • R y is selected from H, halo, CF 3 , C 1-6 alkoxy, and C 3- 6 cycloalkyloxy;
  • each R a is C i-6 alkyl, C 3-7 cycloalkyl, Ci -6 alkoxy, C 3 . 7 cycloalkoxy, halo-Cj-. ⁇ alkyl, CF 3 , ImIo-Ci -6 alkoxy, cyano, halo, thioalkyl, hydroxy , amino, Ci- ⁇ alkylamino, di (Ci- ⁇ ) alkylamino, di (Ci- ⁇ ) alkylamide, carboxyl, (Q- ⁇ ) carboxyester, C ⁇ -6 alkylsulfone, Ci- ⁇ alkylsulfonamide, di (Ci- 6 ) alkyl(alkoxy)amine, C ⁇ -io aryl, C 7 - H alkylaryl, or a 5-7 membered monocyclic heterocycle.
  • the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein X is O.
  • the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein X is O and R' is other than quinoline or isoquinoline.
  • the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein R 4 is C(O)-OR 5 ; wherein R 5 is Ci -9 alkyl.
  • the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein Q is a C 5 ./ 7 saturated or unsaturated chain optionally containing one oxygen atom.
  • Q is a C 5 ./ 7 saturated or unsaturated chain optionally containing one oxygen atom.
  • the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein W is NH-SO 2 -R 2 ; wherein R2 is unsubstituted C3_ 7 cycloalkyl.
  • composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a composition
  • a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a composition
  • a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier, wherein at least one of the additional compounds is an interferon or ribavirin.
  • the present disclosure provides a composition
  • a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier, wherein at least one of the additional compounds is an interferon or ribavirin, wherein the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastiod interferon tau.
  • the present disclosure provides a composition
  • a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier, wherein at least one of the additional compounds is selected from interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti- sense RNA, Imiqimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
  • the present disclosure provides a composition
  • a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier, wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH.
  • a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH.
  • the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof and administering one or two additional compounds prior to, after, or simultaneously with the compound of formula (I), or the therapeutically acceptable salt thereof.
  • at least one of the additional compounds is an interferon or ribavirin.
  • the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A 5 lymphoblastiod interferon tau.
  • the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof and administering one or two additional compounds prior to, after, or simultaneously with the compound of formula (I), or the therapeutically acceptable salt thereof, wherein at least one of the additional compounds is selected from interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'-monophos ⁇ ate dehydrogenase inhibitor, amantadine, and rimantadine.
  • the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof and administering one or two additional compounds prior to, after, or simultaneously with the compound of formula (I), or the therapeutically acceptable salt thereof, wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH.
  • the present disclosure provides macrocyclic isoquinoline compounds of the following formula:
  • R4 is H; alkyl, or C 3-7 cycloalkyl each optionally substituted with halo; alkoxy; -C(O)-R 5 ; C(O)-NHR 5 ; or C(O)-OR 5; wherein R 5 is C 1 .
  • Re is H, Ci -6 alkyl, or C 3-7 cycloalkyl
  • R3 and R' 3 are each independently H or methyl
  • Q is a C3.9 saturated or unsaturated chain, optionally containing one to three heteroatoms independently selected from O or S(O) n ,; wherein m is 0, 1 or 2;
  • W is OH, -O-Rj, or -NH-SO 2 -R 2 : wherein Ri is Ci -6 alkyl, unsaturated C 3-7 cycloalkyl, C 6- I 4 aryl or C 7- I 6 alkylaryl; and R 2 is Ci -8 alkyl, C 4 .
  • alkylcycloalkyl unsubstituted C 3-7 cycloalkyl; or R 2 is cyclopropyl or cyclobutyl optionally substituted with C 1 ⁇ alkyl or C 7 .
  • X is O, S, SO, SO 2 , OCH 2 , CH 2 O or NH;
  • R ! is Het, Ce- 10 aryl or C 7- W alkylaryl, each optionally substituted with R a ; or C3-9 cycloalkyl or Ci- 7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; and (h) R a is Ci-6 alkyl, C 3-7 cycloalkyl, Ci -6 alkoxy, C 3-7 cycloalkoxy, halo-Ci-. 6 alkyl, CF 3 , mono-or di- halo-C
  • compositions comprising the compounds or pharmaceutically acceptable salts, solvates or prodrugs thereof and a pharmaceutically acceptable carrier.
  • the present disclosure provides pharmaceutical compositions useful for inhibiting HCV NS3 protease comprising a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier.
  • the present disclosure further provides methods for treating patients infected with HCV, comprising administering to the patient a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate or prodrug thereof. Additionally, the present disclosure provides methods of inhibiting HCV NS3 protease by contacting the NS3 protease with a compound of the present disclosure.
  • the present disclosure provides peptide compounds that can inhibit the functioning of theMS3 protease, e.g., in combination with the NS4A protease.
  • the present disclosure makes it possible to administer combination therapy to a patient whereby a compound in accordance with the present disclosure, which is effective to inhibit the HCV NS3 protease, can be administered with another compound having anti-HCV activity, e.g., a compound which is effective to inhibit the function of a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH and a nucleoside analog for the treatment of an HCV infection.
  • a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH and a nucleoside analog for the treatment of an HCV infection.
  • racemic mixture and “racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • 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.
  • stereoisomers refers to compounds which have identical chemical composition, but differ with regard to the arrangement of the atoms or groups in space.
  • diastereomer refers to a stereoisomer which is not an enantiomer, e.g., 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, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
  • enantiomers refers to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • pharmaceutically acceptable salt is intended to include nontoxic salts synthesized from a compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • terapéuticaally effective amount means the total amount of each active component that is sufficient to show a meaningful patient benefit, e.g., a sustained reduction in viral load.
  • a meaningful patient benefit e.g., a sustained 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.
  • derivative means a chemically modified compound wherein the modification is considered routine by the ordinary skilled chemist, such as an ester or an amide of an acid, protecting groups, such as a benzyl group for an alcohol or thiol, and tert-butoxycarbonyl group for an amine.
  • solvate means a physical association of a compound of this disclosure with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • solvent encompasses both solution-phase and isolable solvates. Exemplary solvates include hydrates, ethanolates, methanolates, isopropanolates and the like.
  • prodrug means derivatives of the compounds of the disclosure which have chemically or metabolically cleavable groups and become, by solvolysis or under physiological conditions, the compounds of the disclosure which are pharmaceutically active in vivo.
  • a prodrug of a compound may be formed in a conventional manner with a functional group of the compounds such as with an amino, hydroxy or carboxy group when present.
  • the prodrug derivative form often offers advantages of solubility, tissue compatibility, or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985).
  • Prodrugs include acid derivatives well known to practitioners of the art, such as, for example, esters prepared by reaction of the parent acidic compound with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a suitable amine.
  • the term "patient” includes both human and other mammals.
  • composition means a composition comprising a compound of the disclosure in combination with at least one additional pharmaceutical carrier, i.e., adjuvant, excipient or vehicle, such as diluents, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, , perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
  • additional pharmaceutical carrier i.e., adjuents, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, , perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
  • additional pharmaceutical carrier i.e., adjuvant, excipient or vehicle, such as diluent
  • phrases "pharmaceutically acceptable” is employed herein to refer 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 risk/benefit ratio.
  • the term “treating” refers to: (i) preventing a disease, disorder or condition from occurring in a patient which may be predisposed to the disease, disorder and/or condition but has not yet been diagnosed as having it; (ii) inhibiting the disease, disorder or condition, i.e., arresting its development; and (iii) relieving the disease, disorder or condition, i.e., causing regression of the disease, disorder and/or condition.
  • substituted includes substitution at from one to the maximum number of possible binding sites on the core, e.p., organic radical, to which the subsitutent is bonded, e.g., mono-, di-, tri- or tetra- substituted, unless otherwise specifically stated.
  • organic radicals e.g., hydrocarbons and substituted hydrocarbons
  • groups e.g., alkylalkoxyamine or arylalkyl
  • groups include all possible stable configurations, unless otherwise specifically stated. Certain radicals and combinations are defined below for purposes of illustration.
  • halo as used herein means a halogen substituent selected from bromo, chloro, fluoro or iodo.
  • haloalkyl means an alkyl group that in substituted with one or more halo substituents.
  • alkyl as used herein means acyclic, straight or branched chain alkyl substituents having the specified number of carbon atoms and includes, for example, methyl, ethyl, propyl, butyl, tert-butyl, hexyl, 1-methylethyl, 1- methylpropyl, 2-methypropyl, 1,1-dimethylethyl.
  • Ci-e alkyl refers to an alkyl group having from one to six carbon atoms.
  • lower alkyl means an alkyl group having from one to six, preferably from one to four carbon atoms.
  • alkylester means an alkyl group additionally containing on ester group.
  • alkenyl as used herein means an alkyl radical containing at least one double bond, e.g., ethenyl (vinyl) and alkyl.
  • alkoxy as used herein means an alkyl group with the indicated number of carbon atoms attached to an oxygen atom. Alkoxy includes, for example, methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy and 1,1-dimethylethoxy. The latter radical is referred to in the art as tert-butoxy.
  • alkoxycarbonyl means an alkoxy group additionally containing a carbonyl group.
  • cycloalkyl as used herein means a cycloalkyl substituent containing the indicated number of carbon atoms and includes, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and spiro cyclic groups such as spirocyclopropyl as spirocyclobutyl.
  • cycloalkoxy as used herein means a cycloalkyl group linked to an oxygen atom, such as, for example, cyclobutyloxy or cyclopropyloxy.
  • alkylcycloalkyl means a cycloalkyl group linked to an alkyl group.
  • the stated carbon number range includes the total number of carbons in the radical, unless otherwise specfically stated.
  • This a C4-10 alkylcycloalkyl may contain from 1-7 carbon atoms in the alkyl group and from 3-9 carbon atoms in the ring, e.g., cyclopropylmethyl or cyclohexylethyL
  • aryl as used herein means an aromatic moiety containing the indicated number of carbon atoms, such as, but not limited to phenyl, indanyl or naphthyl.
  • C 6- Io aryl refers to an aromatic moiety having from six to ten carbon atoms which may be in the form of a monocyclic or bicyclic structure.
  • haloaryl refers to an aryl mono, di or tri substituted with one or more halogen atoms.
  • alkylaryl arylalkyl
  • aralalkyl mean an aryl group substituted with one or more alkyl groups. Unless the carbon range of each group is specified, the stated range applies to the entire substituent.
  • a C 7 . H alkylaryl group many have from 1-8 carbon atoms in the alkyl group for a monocyclic aromatic and from 1-4 carbon atoms in the alkyl group for a fused aromatic.
  • the attachment of the group to bonding site on the molecule can be either at the aryl group or the alkyl group.
  • aryl radicals include those substituted with typical substituents known to those skilled in the art, e.g., halogen, hydroxy, carboxy, carbonyl, nitro, sulfo, amino, cyano, dialkylamino haloalkyl, CF 3 , haloalkoxy, thioalkyl, alkanoyl, SH, alkylamino, alkylamide, dialkylamide, carboxyester, alkylsulfone, alkylsulfonamide and alkyl(alkoxy)amine.
  • typical substituents e.g., halogen, hydroxy, carboxy, carbonyl, nitro, sulfo, amino, cyano, dialkylamino haloalkyl, CF 3 , haloalkoxy, thioalkyl, alkanoyl, SH, alkylamino, alkylamide, dialkylamide, carboxyester, alkylsulfone, al
  • alkylaryl groups examples include benzyl, butylphenyl and 1-naphthylmethyl.
  • alkanoyl as used herein means straight or branched 1-oxoalkyl radicals containing the indicated number of carbon atoms and includes, for example, formyl, acetyl, 1-oxopropyl (propionyl), 2-methyl-l-oxopropyl, 1-oxohexyl and the like.
  • alkylamide as used herein means an amide mono-substituted with an alkyl, such as
  • heterocycle also referred to as "Het”, as used herein means 7-12 membered bicyclic heterocycles and 5-7 membered monocyclic heterocycles.
  • Preferred bicyclic heterocycles are 7-12 membered fused bicyclic ring systems (both rings share an adjacent pair of atoms) containing from one to four heteroatoms selected from nitrogen, oxygen and sulfur, wherein both rings of the heterocycle are folly unsaturated.
  • the nitrogen and sulfur heteroatoms atoms may be optionally oxidized.
  • the bicyclic heterocycle may contain the heteroatoms in one or both rings.
  • the heterocycles include those substituted with typical substituents known to those skilled in the art.
  • the bicyclic heterocycle may also contain substituents on any of the ring carbon atoms, e.g., one to three substituents.
  • substituents include Cj -6 alkyl, C 3-7 cycloalkyl, Cue alkoxy, C 3-7 cycloalkoxy, halo-Ci- 6 alkyl, CF 3 , mono-or di- halo-Ci- 6 alkoxy, cyano, halo, thioalkyl, hydroxy, alkanoyl, NO 2 , SH, , amino, Ci -6 alkylamino, di (C ⁇ - ⁇ ) alkylamino, di (C ⁇ -e) alkylamide, carboxyl, (Ci- ⁇ ) carboxyester, Ci- ⁇ alkylsulfone, Ci- ⁇ alkylsulfonamide, Ci_ 6 alkylsulfoxide, di (Ci- ⁇ ) alkyl(alkoxy)amine, C ⁇ -ioaryl, C 7-I4 alky laryl, and a 5-7 membered monocyclic heterocycle.
  • bicyclic heterocycles When two substituents are attached to vicinal carbon atoms of the bicyclic heterocycle, they can join to form a ring, e.g., a five, six or seven membered ring system containing up to two heteroatoms selecting from oxygen and nitrogen. Heterocycles may be attached to the parent molecular moiety through any substitutable carbon atom or nitrogen atom in the ring. Examples of bicyclic heterocycles include, but are not limited to, the following ring systems:
  • Preferred bicyclic heterocycles are those other than
  • X and Y together with the carbon atoms to which they are attached, form a ring selected from aryl and heteroaryl;
  • X 1 is selectd from CH and N, and
  • Y 1 and Y 2 are independently selected from CH, O, S, and NH; wherein either ring system can be substituted or unsubstituted at any substitutable carbon or nitrogen atom in the ring.
  • Preferred monocyclic heterocycles are 5-7 membered saturated, partially saturated or fully unsaturated ring system (this latter subset also herein referred to as unsaturated heteroaromatic) containing in the ring from one to four heteroatoms selected from nitrogen; oxygen and sulfur, wherein the sulfur and nitrogen heteroatoms may be optionally oxidized.
  • unsaturated heteroaromatic 5-7 membered saturated, partially saturated or fully unsaturated ring system
  • the heterocycles include those substituted with typical substituents known to those skilled in the art.
  • the monocyclic heterocycle may also contain substituents on any of the ring atoms, e.g., one to three substituents.
  • suitable substituents include Ci -6 alkyl, C 3-7 cycloalkyl, Ci_6 alkoxy, C3-7 cycloalkoxy, halo-Ci-g alkyl, CF3, mono-or di- halo-Ci- 6 alkoxy, cyano, halo, thioalkyl, hydroxy, alkanoyl, NO 2 , SH, amino, Ci- ⁇ alkylamino, di (Ci- ⁇ ) alkylamino, di (Cj-g) alkylamide, carboxyl, (Ci-e) carboxyester, Cj -6 alkylsulfone, C 1 -6 alkylsulfoxide, C 1.
  • the monocyclic heterocycle may be attached to the molecule, e.g. Ri in formula (I), at any atom in the ring.
  • monocyclic heterocycles include, but are not limited to, the following (and their tautomers):
  • heterocycles used in the compounds of the present disclosure should be stable.
  • stable compounds are those which can be synthesized, isolated and formulated using techniques known the those skilled in the art without degradation of the compound.
  • substituted with reference to an amino acid or amino acid derivative means a radical derived from the corresponding ⁇ -amino acid.
  • substituents methyl, iso-propyl, and phenyl represent the amino acids alanine, valine, and phenyl glycine, respectively.
  • Pl', Pl, P2, P3 and P4" map the relative positions of the amino acid residues of a protease inhibitor binding relative to the binding of the natural peptide cleavage substrate. Cleavage occurs in the natural substrate between Pl and Pl ' where the nonprime positions designate amino acids starting from the C-terminus end of the peptide natural cleavage site extending towards the N-terminus; whereas, the prime positions emanate from the N-terminus end of the cleavage site designation and extend towards the C-terminus.
  • Pl ' refers to the first position away from the right hand end of the C-terminus of the cleavage site (ie.
  • R 4 is H; C 1 - 6 alkyl, or C 3-7 cycloalkyl each optionally substituted with halo; alkoxy; -C(O)-R 5 ; C(O)-NHR 5 ; or C(O)-OR 5 ; wherein R 5 is Ci -9 alkyl, optionally substituted with Ci. 6 alkoxy, € 3 . 7 cycloalkoxy, halo-Ci- 6 alkoxy, cyano, halo, hydroxy, amino, Ci.
  • R6 is H, Cl-6 alkyl, or C3-7 cycloalkyl;
  • R3 and R'3 are each independently H or methyl;
  • Q is a C3-9 saturated or unsaturated chain, optionally containing one to three heteroatoms independently selected from O or S(O)m; wherein m is 0, 1 or 2;
  • W is OH, -O-Rl , or -NH-SO2-R2: wherein Rl is Cl -6 alkyl, unsaturated C3- 7 cycloalkyl, C6-14 aryl or C7-16 alkylaryl; and R2 is Cl-8 alkyl, C4-10 alkylcycloalkyl, unsubstituted C3-7 cycloalkyl; or R2 is cyclopropyl or cyclobutyl optionally substituted with C 1-4 alkyl or C7-16 alkylaryl, alkoxy, halo, haloalkyl, cyano, alkylcyano, or haloalkoxy;
  • X is O, S 5 SO, SO2, OCH2, CH2O or NH;
  • R' is Het, C6-10 aryl or C7-14 alkylaryl, each optionally substituted with Ra; or C3-9 cycloalkyl or C 1-7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; and
  • Ra is Cl-6 alkyl, C3-7 cycloalkyl, Cl-6 alkoxy, C3-7 cycloalkoxy, halo-Cl-6 alkyl, CF3, mono-or di- halo-Cl-6 alkoxy, cyano, halo, thioalkyl, hydroxy, alkanoyl, NO2, SH, , amino, Cl-6 alkylamino, di (Cl-6) alkylamino, di (Cl-6) alkylamide, carboxyl, (Cl-6) carboxyester, Cl-6 alkylsulfone, Cl-6 alkylsulfonamide, di (Cl-6) alkyl(alkoxy)amine, C6-10 aryl, C7-14 alkylaryl, or a 5-7 membered monocyclic heterocycle.
  • X is O 5 S, OCH 2 , CH 2 O or NH. In another aspect, X is O or OCH 2 . In another aspect, X is O. In one aspect, R' is Het or C 6-1O aryl, each optionally substituted with R a . In another aspect, R' is Het. Often, the Het contains 1 or 2 nitrogen atoms and optionally a sulfur atom or an oxygen atom in the ring. In one aspect, the heterocycle is substituted with at least one of Ci -6 alkyl, Ci .6 alkoxy, halo, Ce aryl, and a 5-7 membered monocyclic heterocycle.
  • R' is a bicyclic heterocycle containing 1 nitrogen atom and substituted with methoxy and at least one of a Ce aryl and a 5-7 membered monocyclic heterocycle.
  • R' is a monocyclic heterocycle containing 1 or 2 nitrogen atoms and substituted with methoxy and at least one of a Ce aryl and a 5-7 membered monocyclic heterocycle.
  • R' has a structure selected from:
  • R' has a structure selected from:
  • R' is selected from the following heterocycles
  • X-IT is selected from the following;
  • Ra is C 1-6 alkyl, C 3-7 cycloalkyl, C 1-6 alkoxy, C 3-7 cycloalkoxy, halo-C 1 _ 6 alkyl, CF 3 , halo-Cue alkoxy, cyano, halo, thioalkyl, hydroxy , amino, Ci .
  • R a is C 1-6 alkyl, C 3-7 cycloalkyl, C 1-6 alkoxy, C3 -7 cycloalkoxy, halo-C 1-.6 alkyl, CF 3 , halo-Ci- 6 alkoxy, cyano, halo, di (C 1 - O ) alkylamino, di (C 1-6 ) alkylamide, C 6- Io aryl, C 7 . 14 alkylaryl, or a 5-7 membered monocyclic heterocycle.
  • R a is C L6 alkyl, C ⁇ . ⁇ cycloalkyl, Ci ⁇ alkoxy, Ca -7 cycloalkoxy, halo-Ci-.
  • R a is Ci- 6 alkyl, C 3 . 7 cycloalkyl, C 1-6 alkoxy, halo-Ci- 6 alkyl, halo, amino, C ⁇ aryl, or a 5- 7 membered monocyclic heterocycle.
  • R a is Ci- 6 alkoxy or halo.
  • R a is methoxy or chloro.
  • -X-R' has a structure other than the following structure:
  • R 7 is H, halo, Cl-6 alkyl, Cl-6 alkoxy, C3-6 cycloalkoxy, phenyl optionally substituted with Cl -3 alkyl, C 1-3 alkoxy or halo, or heterocycle optionally substituted with from one to three of halo, cyano, trifluoromethyl, nitro, Cl-6 alkyl, Cl-6 alkoxy, amido, Cl-6 alkanoylamino or amino; and
  • R 8 is H, halo, CF3, Cl-6 alkoxy, C3-6 cycloalkoxy or C 1-3 alkyl.
  • W is -NH-SO 2 -R 2 : wherein R 2 is C 1-8 alkyl, C 4-10 alkylcycloalkyl, unsubstituted C 3-7 cycloalkyl; or R 2 is cyclopropyl or cyclobutyl optionally substituted with Ci -4 alkyl or C 7- i 6 alkylaryl.
  • R 2 is Ci -8 alkyl, C 4- io alkylcycloalkyl or unsubstituted C 3-7 cycloalkyl.
  • R 2 is cyclopropyl or cyclobutyl.
  • Q is a Cs -7 saturated or unsaturated chain optionally containing one to three heteroatoms independently selected from O or S(O) n ,; wherein m is 0, 1 or 2.
  • Q is a Ce saturated or unsaturated chain optionally containing one to three heteroatoms independently selected from O or S(O) m ; wherein m is 0, 1 or 2.
  • Q is unsaturated.
  • Q has six carbon atoms.
  • Q has a structure selected from:
  • P is a C3 saturated chain optionally containing one heteroatom independently selected from O, S(0)m; wherein m is 0, 1 or 2.
  • R 2 is cyclopropyl substituted in the Cl position with C 1-4 alkyl or C7-16 alkylaryl, or the C2 or C3 position with Cl-4 alkyl.
  • R 2 is cyclobutyl substituted in the Ci position with C 1-4 alkyl or C 7- 1 ⁇ alkylaryl, or the C 2 , C3 or C 4 position with C M alkyl.
  • R 4 is -C(O)-R 5 , C(O)-NHR 5 or C(O)- OR 5 ; wherein R 5 is Ci -6 alkyl optionally substituted with halo, alkoxy, or cyano.
  • R 5 is Cj. 6 alkyl optionally substituted with halo.
  • R 5 is Ci-6 alkyl.
  • R 3 and R' 3 are each independently H or methyl.
  • R 6 is H or Ci -6 alkyl.
  • the compounds of the present disclosure have the structure of formula (II):
  • R 4 is -C(O)-R 5 , C(O)-NHR 5 or C(O)-OR 5; wherein R 5 is C ]-9 alkyl optionally substituted with Ci -6 alkoxy, cyano, halo;
  • Q is a C 5-7 saturated or unsaturated chain optionally containing one to three heteroatoms independently selected from O or S(O) m ; wherein m is 0, 1 or 2;
  • W is NH-SO2-R 2 , wherein R 2 is Ci-8 alkyl, C 4- io alkylcycloalkyl, unsubstituted C 3 . 7 cycloalkyl, or cyclopropyl or cyclobutyl optionally substituted with Ci -4 alkyl or C7-16 alkylaryl;
  • X is O, S, OCH 2 , CH 2 O or NH;
  • R' is Het, C ⁇ -io aryl or C 7-H alkylaryl, each optionally substituted with R a ; or C 3 . 9 cycloalkyl or C 1 . 7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; and (f) R a is Ci-6 alkyl, C 3 . 7 cycloalkyl, C1.6 alkoxy, C 3-7 cycloalkoxy, halo-Ci-.g alkyl, CF 3 , halo-Ci.
  • R 4 is -C(O)-R 5 , C(O)-NHR 5 or C(O)-OR 5 ,- wherein R 5 is Ci -9 alkyl optionally substituted halo;
  • Q has a structure selected from;
  • P is a C 3 saturated chain optionally containing one heteroatom independently selected from O, S(O) n ,; wherein m is 0, 1 or 2;
  • W is NH-SO 2 -R 2 , wherein R 2 is C 1-8 alkyl, C 3 -? cycloalkyl,
  • X is O, or OCH 2 ;
  • R' is Het, C ⁇ -io ary.1 or C 7-I4 alkylaryl, each optionally substituted with R a ; and R a is C 1-6 alkyl, C 3-7 cycloalkyl, C ⁇ alkoxy, C 3-7 cycloalkoxy, halo-Ci-. ⁇ alkyl, halo- Ci -6 alkoxy, cyano, halo, di (Q-e) alkylamino.
  • R 4 is -C(O)-R 5 , C(O)-NHR 5 or C(O)-OR 5 ; wherein R 5 is Ci -9 alkyl;
  • W is NH-SO 2 -R 2 , wherein R 2 is cyclopropyl
  • R' is Het or C ⁇ -io aryl each optionally substituted with R a ; and R a is methoxy or chloro.
  • the compounds of the present disclosure have the structure of formula (III):
  • R 4 is -C(O)-R 5 , C(O)-NHR 5 or C(O)-OR 5 ; wherein R 5 is C 1-6 alkyl;
  • W is NH-SO 2 -R 2 , wherein R2 is cyclopropyl; and (d) R' is Het or Ce- 10 aryl.
  • Q has a structure selected from:
  • R' has a structure selected from the following
  • each R' is optionally substituted with R a ; wherein R a is selected from alkoxy, c.yano, cycloalkoxy, halo, or dialkyamino.
  • R 1 has a structure selected from the following
  • each R' is optionally substituted with R a ; wherein R a is selected from alkoxy or halo.
  • the compounds of the present disclosure which contain a basic moiety, can form salts by the addition of a pharmaceutically acceptable acid.
  • the acid addition salts are formed from a compound of formula (I) and a pharmaceutically acceptable inorganic acid, including but not limited to hydrochloric, hydrobromic, hydroiodic, sulfuric, phosphoric, or organic acid such as/?-toluenesulfonic, methanesulfonic, acetic, benzoic, citric, malonic, fumaric, maleic, oxalic, succinic, sulfamic, or tartaric.
  • examples of such pharmaceutically acceptable salts include chloride, bromide, iodide, sulfate, phosphate, methanesulfonate, citrate, acetate, malonate, fumarate, sulfamate, and tartrate.
  • Salts of an amine group may also comprise quaternary ammonium salts in which the amino nitrogen carries a suitable organic group such as an alkyl, alkenyl, alkynyl or aralkyl moiety.
  • Compounds of the present disclosure, which are substituted with an acidic group may exist as salts formed through base addition.
  • Such base addition salts include those derived from inorganic bases which include, for example, alkali metal salts (e.g. sodium and potassium), alkaline earth metal salts (e.g. calcium and magnesium), aluminum salts and ammonium salts.
  • suitable base addition salts include salts of physiologically acceptable organic bases such as trimethylamine, triethylamine, morpholine, pyridine, piperidine, picoline, dicyclohexylamine, N,N' -dibenzylethylenediamine, 2-hydroxyethylamine, bis-(2-hydroxyethyl)amine, tri-(2-hydroxyethyl)amine, procaine, dibenzylpiperidine, N-benzyl- ⁇ -phenethylamine, dehydroabietylamine, N 5 N '-bishydroabietylamine, glucamine, N-methylglucamine, collidine, quinine, quinoline, ethylenediamine, ornithine, choline, N,N'-benzylphenethylamine, chloroprocaine, diethanolamine, diethylamine, piperazine, tris(hydroxymethyl)aminomethane and tetramethylammonium hydroxide and basic amino
  • Certain compounds of the present disclosure, and their salts may also exist in the form of solvates with water, for example hydrates, or with organic solvents such as methanol, ethanol or acetonitrile to form, respectively, a methanolate, ethanolate or acetonitrilate.
  • the present disclosure includes each solvate and mixtures thereof.
  • compounds of the present disclosure, or a salt or solvate thereof may exhibit polymorphism.
  • the present disclosure also encompasses any such polymorphic form.
  • Asymmetric centers exist in the compounds of the present disclosure.
  • the compounds may include Pl cyclopropyl element of formula
  • Ci and C 2 each represent an asymmetric carbon atom at positions 1 and 2 of the cyclopropyl ring. Not withstanding other possible asymmetric centers at other segments of the compounds, the presence of these two asymmetric centers means that the compounds can exist as racemic mixtures of diastereomers, such as the diastereomers wherein R 2 is configured either syn to the amide or syn to the carbonyl as shown below.
  • the disclosure encompasses all stereochemical isomeric forms, or mixtures thereof, which possess the ability to inhibit HCV protease.
  • the enantiomers may be resolved by methods known to those skilled in the art, for example, by formation of diastereoisomeric salts which may be separated by crystallization, gas-liquid or liquid chromatography, selective reaction of one enantiomer with an enantiomer-specific.reagent. It will be appreciated that where the desired enantiomer is converted into another chemical entity by a separation technique, then an additional step is required to form the desired enantiomeric form.
  • specific enantiomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer into the other by asymmetric transformation.
  • Certain compounds of the present disclosure may also exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers.
  • the present disclosure includes each conformational isomer of these compounds and mixtures thereof.
  • Certain compounds of the present disclosure may exist in zwitterionic form and the present disclosure includes each zwitterionic form of these compounds and mixtures thereof.
  • the compounds of the present disclosure can be manufactured by methods known to those skilled in the art. The following methods set forth below are provided for illustrative purposes and are not intended to limit the scope of the claimed disclosure.
  • compounds of the present disclosure having the structure of formula (I) can be synthesized, as shown in scheme 1. It will be recognized that it may be preferred or necessary to prepare such a compound in which a functional group is protected using a conventional protecting group then to remove the protecting group to provide a compound of the present disclosure.
  • the details concerning the use of protecting groups in accordance with the present disclosure are known to those skilled in the art.
  • intermediates of the present disclosure such as dipeptide 1
  • the Boc protected nitrogen of 1 is deprotected using an acid such as HCl in a solvent such as ether, to provide the corresponding free amine 2.
  • Amine 2 can be subsequently coupled to amino acid 3 using a coupling agent such as HATU in a solvent such as dichloromethane to provide the tripeptide intermediate 4.
  • a coupling agent such as HATU in a solvent such as dichloromethane
  • a key transformation in the construction of compounds of formula (I) is the macrocyclization process wherein intermediates of general structure 4 are converted into intermediates of general structure 5.
  • the conversion of intermediate 4 into 5 can be affected by an intramolecular olefin metathesis reaction.
  • This class of reactions is well established in the art and as such, a number of olefln-metathesis-catalysts have been developed and are commercially available.
  • the conversion of diene 4 to macrocycle 5 could be affected by the treatment of 4 with a sufficient quantity of Grubb's first-generation olefin metathesis catalyst, in a solvent such as dichloromethane or dichloroethane.
  • Intermediate 5 is then coverted to compounds of formula (I) such as 7 by a two step process.
  • the ester functionality of intermediate 5 is hydrolyzed to the corresponding carboxylic 6.
  • This transformation can be accomplished by a saponification reaction wherein 5 is treated with a base such as lithium hydroxide in a mixture of THF 5 methanol and water.
  • the resulting acid 6 can be converted to a compound of formula (I) by a simple coupling reaction with a sulfonamide derivative as shown.
  • the PT terminus is incorporated into the molecules using one of the general processes outlined above and described in more detail below.
  • the P 1 " elements that is the cycloalkylsulfonamides or alkyl sulfonamides, are commercially available or can be prepared from the corresponding alkyl- or cycloalkyl-sulfonyl chloride by treating said sulfonyl chloride with ammonia .
  • these sulfonamides can be synthesized using the general process outlined in Scheme 3.
  • 3-chloropropylsulfonyl chloride (1) is converted to a suitable protected sulfonamide as for example by treatment with tert-butyl amine.
  • the sulfonamide 2 obtained is then converted to the corresponding cycloalkylsulfonamide 3 by treatment with two equivalents of a base such as butyl lithium in a solvent such as THF at low temperature.
  • the resulting cycloalkylsulfonamide can be deprotected by treatment with an acid to provide the desired unprotected cycloalkylsulfonamide 4.
  • Said PV fragment 4 can be incorporated into compounds of formula (I). Additionally, the cycloalkyl ring of intermediates like 4 can be further functionalized.
  • intermediate 3 treatment of intermediate 3 with a base such as butyl lithium followed by the addition of an electrophile such as an alkyl halide should provide intermediates like 5, wherein the Cl position of the cycloalkyl ring is functionalized.
  • Reactions of this type can be conducted in solvents such as THF. In such a reaction it may be necessary to add two or more equivalents of base to intermediate 3. Moreover, the temperature of such a reaction will likely need to be carefully monitored wherein the THF solution of 3 is cooled to -78C prior to the addition of base and this is described in detail herein.
  • a Boc group can be employed as shown below (Scheme 4).
  • Said Boc group can be incorporated by treatment of an intermediate like 2 with Boc anhydride in the presence of a base such as triethylamine in conjunction with catalytic DMAP.
  • the acylsulfonamide 3 obtained is then converted to the corresponding cycloalkylacylsulfonamide 4 by treatment with two equivalents of a base such as butyl lithium in a solvent such as THF at low temperature.
  • the resulting cycloalkylacylsulfonamide 4 can be deprotected by treatment with an acid to provide the desired unprotected cycloalkylsulfoamide.
  • Said PT fragment can be incorporated into compounds of formula (I).
  • dipeptide intermediates like 2 shown below can be prepared by the coupling of hydroxyproline derivative 1 with cyclopropyl amino acid B as shown. This coupling reaction can be carried out using reagents such as HATU or HBTU and in solvents such as methylene chloride or DMF or THF.
  • removal of the Boc group from intermediate 4a can be accomplished by subjecting 4a to an acid such as HCl in a solvent such as ether, to provide the corresponding amine hydrochloride 6.
  • Intermediate 6 can then be coupled to a functionalized proline moiety 1 to provide the P1-P2 dipeptide 2.
  • Intermediates like 2 can be converted to compounds of formula (I) by the methods described herein.
  • the resulting amine TFA salt can be treated with an isocyanate in the presence of one equivalent of base to provide a compound of formula (I) (3) wherein the P3 moiety is capped with a urea.
  • intermediate 2 can be used as starting material for the preparation of compounds of formula (I) wherein the P3 group is capped with an amide or a carbamate.
  • the construction of said compounds of formula (I) can be achieved using standard conditions for the formation of said P4 functionalities from amines.
  • compositions comprising a compound of the present disclosure, or a pharmaceutically acceptable enantiomer, diastereomer, salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier.
  • Pharmaceutical compositions of the present disclosure comprise a therapeutically effective amount of a compound of the disclosure, or a pharmaceutically acceptable enantiomer, diastereomer, salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier, with a pharmaceutically acceptable carrier, e.g., excipient, or vehicle diluent.
  • the active ingredient, i.e., compound, in such compositions typically comprises from 0.1 weight percent to 99.9 percent by weight of the composition, and often comprises from about 5 to 95 weight percent.
  • a composition comprising the compound of formula (I) and a pharmaceutically acceptable carrier.
  • the composition further comprises a compound having anti-HCV activity.
  • anti-HCV activity means the compound is effective to inhibit the function of a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH and a nucleoside analog for the treatment of an HCV infection.
  • the other compound having anti-HCV activity is effective to inhibit the function of target in the HCV life cycle other than the HCV NS3 protease protein.
  • the compound having anti-HCV activity is an interferon.
  • the interferon is selected from the group consisting of interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, lymphoblastiod interferon tau.
  • the compound having anti-HCV activity is selected from the group consisting of interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
  • the composition comprises a compound of the disclosure, an interferon and ribavirin.
  • the compound having anti-HCV activity is a small molecule compound.
  • small molecule compound means a compound having a molecular weight of less than 1,500 daltons, preferably less than 1000 daltons.
  • the small molecule compound is effective to inhibit the function of a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, inosine monophophate dehydrogenase ("IMPDH”) and a nucleoside analog for the treatment of an HCV infection.
  • a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, inosine monophophate dehydrogenase (“IMPDH”) and a nucleoside analog for the treatment of an HCV infection.
  • IMPDH inosine monophophate dehydrogenase
  • Table 1 lists some illustrative examples of compounds that can be administered with the compounds of this disclosure.
  • the compounds of the disclosure can be administered with other anti-HCV activity compounds in combination therapy, either jointly or separately, or by combining the compounds into a composition.
  • compositions of this disclosure may be administered orally, parenterally or via an implanted reservoir. Oral administration or administration by injection are preferred. In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, and intralesional injection or infusion techniques.
  • the pharmaceutical compositions of this disclosure may be administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents, such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • suitable carriers for the above noted compositions can be found in standard pharmaceutical texts, e.g. in "Remington's Pharmaceutical Sciences", 19th ed., Mack Publishing Company, Easton, Perm., 1995.
  • compositions of this disclosure can be prepared by known procedures using well-known and readily available ingredients.
  • the compositions of this disclosure may be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art.
  • the active ingredient will usually be admixed with a carrier, or diluted by a carrier, or enclosed within a carrier which may be in the form of a capsule, sachet, paper or other container.
  • the carrier serves as a diluent, it may be a solid, semi-solid or liquid material which acts as a vehicle, excipient or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, beadlets, lozenges, sachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols, (as a solid or in a liquid medium), soft and hard gelatin capsules, suppositories, sterile injectable solutions, sterile packaged powders and the like. Further details concerning the design and preparation of suitable delivery forms of the pharmaceutical compositions of the disclosure are known to those skilled in the art.
  • Dosage levels of between about 0.01 and about 1000 milligram per kilogram (“mg/kg”) body weight per day, preferably between about 0.5 and about 250 mg/kg body weight per day of the compounds of the disclosure are typical in a monotherapy for the prevention and treatment of HCV mediated disease.
  • 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.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. As the skilled artisan will appreciate, lower or higher doses than those recited above may be required.
  • Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the infection, the patient's disposition to the infection and the judgment of the treating physician.
  • treatment is initiated with small dosages substantially less than the optimum dose of the peptide. 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 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 100%, and more preferably between about 10 and 80% of the dosage normally administered in a monotherapy regimen.
  • the resulting composition may be administered in vivo to mammals, such as man, to inhibit HCV NS 3 protease or to treat or prevent HCV virus infection.
  • another aspect of this disclosure provides methods of inhibiting HCV NS3 protease activity in patients by administering a compound of the present disclosure or a pharmaceutically acceptable enantiomer, diastereomer, salt or solvate thereof.
  • a method of treating an HCV infection in a patient comprising administering to the patient a therapeutically effective amount of the compound of the disclosure, or a pharmaceutically acceptable enantiomer, diastereomer, solvate, prodrug or salt thereof.
  • the method of administering the compound is effective to inhibit the function of the HCV NS3 protease protein, hi a preferred aspect, the method further comprises administering another compound having anti-HCV activity (as described above) prior to, after or concurrently with a compound of the disclosure.
  • the compounds of the disclosure may also be used as laboratory reagents. Compounds may be instrumental in providing research tools for designing of viral replication assays, validation of animal assay systems and structural biology studies to further enhance knowledge of the HCV disease mechanisms. Further, the compounds of the present disclosure are useful in establishing or determining the binding site of other antiviral compounds, for example, by competitive inhibition.
  • the compounds of this disclosure may also be used to treat or prevent viral contamination of materials and therefore reduce the risk of viral infection of laboratory or medical personnel or patients who come in contact with such materials, e.g., blood, tissue, surgical instruments and garments, laboratory instruments and garments, and blood collection or transfusion apparatuses and materials.
  • materials e.g., blood, tissue, surgical instruments and garments, laboratory instruments and garments, and blood collection or transfusion apparatuses and materials.
  • compositions of the disclosure can be used for the manufacture of a medicament for treating HCV infection in a patient.
  • Solution percentages express a weight to volume relationship, and solution ratios express a volume to volume relationship, unless stated otherwise.
  • Nuclear magnetic resonance (NMR) spectra were recorded either on a Bruker 300, 400 or 500 megahertz (MHz) spectrometer; the chemical shifts ( ⁇ ) are reported in parts per million. Flash chromatography was carried out on silica gel (SiO 2 ) according to Still's flash chromatography technique (J. Org. Chem. 1978, 43, 2923).
  • LC Liquid Chromatography
  • MS Mass Spectrometry
  • Solution percentages express a weight to volume relationship
  • solution ratios express a volume to volume relationship, unless stated otherwise.
  • Nuclear magnetic resonance (NMR) spectra were recorded either on a Bruker 300, 400 or 500 MHz spectrometer; the chemical shifts ( ⁇ ) are reported in parts per million.
  • Example 1 The examples, compounds and chemical intermediates of the present disclosure, described in the following examples, were prepared according to the following methods.
  • Example 1 Example 1:
  • the named compound was made racemic by each of the following methods A and B.
  • Glycine ethyl ester hydrochloride (303.8 g, 2.16 mole) was suspended in tert- butylmethyl ether (1.6 L). Benzaldehyde (231 g, 2.16 mole) and anhydrous sodium sulfate (154.6 g, 1.09 mole) were added and the mixture cooled to 0 °C using an ice- water bath. Triethylamine (455 mL, 3.26 mole) was added dropwise over 30 min and the mixture stirred for 48 h at rt. The reaction was then quenched by addition of ice- cold water (1 L) and the organic layer was separated.
  • the enantio-excess of the ester was determined to be 97.2%, and the reaction was cooled to room temperature (26°C) and stirred overnight (16h) after which the enantio- excess of the ester was determined to be 100%.
  • the pH of the reaction mixture was then adjusted to 8.5 with 50% NaOH and the resulting mixture was extracted with MTBE (2 x 2 L).
  • the combined MTBE extract was then washed with 5% NaHCO 3 (3 x 100 mL), water (3 x 100 mL), and evaporated in vacuo to give the enantiomerically pure N-Boc-(li2,2S)/-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester as light yellow solid (42.55 g; purity: 97% @ 210 nm, containing no acid; 100% enantiomeric excess ("ee").
  • the aqueous layer from the extraction process was then acidified to pH 2 with 50% H 2 SO 4 and extracted with MTBE (2 x 2 L).
  • the MTBE extract was washed with water (3 x 100 mL) and evaporated to give the acid as light yellow solid (42.74 g; purity: 99% @ 210 nm, containing no ester).
  • enantio-excess of the ester was determined to be 44.3% as following: 0.1 mL of the reaction mixture was removed and mixed well with 1 mL ethanol; after centrifugation, 10 microliter (" ⁇ l") of the supernatant was analyzed with the chiral HPLC. To the remaining reaction mixture, 0.1 mL of DMSO was added, and the plate was incubated for additional 3 days at 250 rpm at 40 0 C, after which four mL of ethanol was added to the well. After centrifugation, 10 ⁇ l of the supernatant was analyzed with the chiral HPLC and enantio-excess of the ester was determined to be 100%. Resolution C
  • enantio-excess of the ester was determined to be 39.6% as following: 0.1 mL of the reaction mixture was removed and mixed well with 1 mL ethanol; after cenrifugation, 10 ⁇ l of the supernatant was analyzed with the chiral HPLC. To the remaining reaction mixture, 0.1 mL of DMSO was added, and the plate was incubated for additional 3 days at 250 rpm at 40 0 C, after which four mL of ethanol was added to the well. After centrifugation, 10 ⁇ l of the supernatant was analyzed with the chiral HPLC and enantio-excess of the ester was determined to be 100%. Sample analysis was carried out in the following manner: 1) Sample preparation: About 0.5 ml of the reaction mixture was mixed well with 10 volume of EtOH. After centrifugation, 10 ⁇ l of the supernatant was injected onto HPLC column. 2) Conversion determination:
  • reaction temperature was then adjusted to 4O 0 C. After 3 hours, pH was adjusted to 8.0 with IO N NaOH. After 21 hours, the reaction was cooled down to 25 0 C. pH of the reaction mixture was adjusted to 8.5 with IO N NaOH and the mixture was extracted with MTBE (2 x 5 L). The combined MTBE extract was washed with 5% NaHCO 3 (3 x 500 ml) and water (3 x 200 ml), and evaporated to give 110 gram of yellow oil.
  • the crystal structure enantiomerically pure iV-Boc-(l/?,2,S)/-l-amino-2- vinylcyclopropane carboxylic acid ethyl ester has been characterized by single crystal analysis (X-ray NB#: 52795-093, refcode: 634592N1). The absolute configuration is not established for lack of a known chiral center or heavier atom(s). A chain structure along the crystallographic ⁇ -axis is formed via intermolecular hydrogen bonding between the amide group and the carbonyl oxygen atom (N...O 3.159 A). Structure of iV-Boc-(li?,2iS)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester:
  • Step 1 Preparation of ethyl l(R)-amino-2(S)-vinylcyclopropane carboxylate hydrochloride
  • Ethyl l(R)-tert-butoxycarbonylamino-2(S)-vinylcyclopropanecarboxylate (8.5 g, 33.3 mmol) was stirred under an N 2 atmosphere with 200 mL of 4N HCl/dioxane (Aldrich) at rt for 3h. The solvent was removed under reduced pressure keeping the temperature below 40 C. This gave 6.57 g (-100%) of ethyl l(R)-amino-2(S)- vinylcyclopropanecarboxylate hydrochloride as a light tan solid.
  • Step 2 Preparation of ethyl 1 (R)-[I -tert-butoxycarbonyl-4 (R)-hydroxypyrrolidine- 2(S)-carboxamido]-2(S)-vinylcyclopropanecarboxylate
  • Step 3 Preparation of ethyl l(R)-[4(R)-hydroxypyrrolidine-2(S)-carboxamido]-2(S)- vinylcyclopropanecarboxylate hydrochloride.
  • tert-Butylamine (3.0 mol, 315.3 tnL) was dissolved in THF (2.5 L). The solution was cooled to — 20 0 C. 3-Chloropropanesulfonyl chloride (1.5 mol, 182.4 mL) was added slowly. The reaction mixture was allowed to warm to rt and stirred for 24 h. The mixture was filtered, and the filtrate was concentrated in vacuo. The residue was dissolved in CH2CI2 (2.0 L). The resulting solution was washed with 1 N HCl (1.0 L), water (1.0 L), brine (1.0 L) and dried over Na 2 SO 4 . It was filtered and concentrated in vacuo to give a slightly yellow solid, which was crystallized from hexane to afford the product as a white solid (316.0 g, 99%).
  • Step Ib Preparation ofN-tert-Butyl-(l-methyl)cyclopropylsulfonamide.
  • reaction mixture was warmed to rt, recooled to -78 0 C over a period of 2 h and a neat solution of methyl iodide (5.68 g, 40 mmol) added.
  • the reaction mixture was allowed to warm to rt overnight, quenched with saturated NH 4 Cl (10O mL) at rt. It was extracted with EtOAc (100 mL).
  • Steps Ib Preparation ofN-tert-Butyl- ⁇ l-benzytycyclopropyl-sulfonamide.
  • Steps Ib Preparation of N-tert-Butyl-(l -benzyl) cyclopropyl-sulfonamide.
  • reaction mixture was allowed to warm to room temperature, stirred an additional 3 hours and was pardoned with IN HCl (300 mL), water (300 mL), brine (300 mL), dried over MgSO4, filtered, and concentrated in vacuo to afford the crude product.
  • Step 1 Preparation ofl-methoxymethylcyclopropylsulfonylamine tert- butylcarbamate
  • Step 1 Preparation of 1-cyclopropylmethylcyclopropylsulfonylamine tert- - butylcarbamate
  • This compound was obtained in 65% yield from 1- cyclopropylmethylcyclopropylsulfonylamine terf-butylcarbamate according to the procedure described for the synthesis of l-methoxymethylcyclopropylsulfonamide.
  • Step 1 Preparation of l-propylcarbamoylcyclopropanesulfonamide tert- butylcarbamate
  • Step 1 Preparation of l-(3,5-dimethylisoxazol-4- yl)carbamoylcyclopropanesulfonamide tert-butylcarbamate
  • Step 2 Preparation of l-(3,5-dimethylisoxazol-4- yljcarbamoylcyclopropanesulfonamide
  • Step 1 Preparation of 1 (R)-tert-butoxycarbonylamino-2(S)-vinyl- cyclopropanecarboxylic acid
  • Step 2 Preparation ofl- ⁇ [l-(2(S)-tert-Butoxycarbonylamino-non-8-enoyl)-4(R)- hydroxy-pyrrolidine-2(S)carbonyl]-(lR)-amino ⁇ -2(S)-vinyl-cyclopropanecarboxylic acid ethyl ester '
  • Step 3 Preparation of(lS,4R,6S,14S,18R)-7-cis-14-tert-butoxycarbonylamino- 18-hydroxy-2, 15-dioxo-3, 16-diazatricyclo[l 4.3.0.0 4>6 ]nonadec- 7-ene-4-carboxylic acid ethyl ester
  • Step 4 (IS, 4R, 6SJ4S, 18R)-7-cis-14-tert-butoxycarbonylamino-18-hydroxy-2, 15- dioxo-3, 16-diazatricyclofI 4.3.0.0 4 ' 6 ]-nonadec- 7-ene-4-carboxylic acid
  • Example 18 (1 S,4R,6S, 14S, 1 SR)-I -cis- 14-tert-butoxycarbonylamino- 18-hydroxy-2, 15-dioxo- 3,16-diazatricyclo[14.3.0.0 4 ' 6 ]-nonadec-7-ene-4-carboxylic acid as a gray solid.
  • Step 1 Preparation ofl- ⁇ [l-(2-tert-Butoxycarbonylamino-non-8-enoyl)-4-(tert- butyl-dimethyl-silanyloxy)-pyrrolidine-2-carbonyl]-amino ⁇ -2- vinylcyclopropanecarboxylic acid ethyl ester
  • Step 2 Preparation ofl4-tert-Butoxycarbonylamino-18-(tert-butyl-dimethyl- silanyloxy)-2,l 5-dioxo-3, 16-diaza-tricyclo[14.3.0.0 4 ' 6 ]nonadec-7-ene-4-carboxylic acid, ethyl ester
  • Step 3 Preparation ofl4-tert-butoxycarbonylamino-18-(tert-butyl-dimethyl- silanyloxy)-2,l 5-dioxo-3, 16-diaza-tricyclo[l 4.3.0.0 4 ' 6 ]nonadec- 7-ene-4-carboxylic acid
  • Step 4 Preparation of[18-(tert-butyl-dimethyl-silanyloxy)-4- cyclopropanesulfqnylam inocar bony 1-2, 15-dioxo-3, 16-dia ⁇ a- tricyclo[14.3.0.0 4 ' 6 ]nonadec-7-en-14-yl]-carbamic acid tert-butyl ester
  • Step 5 Preparation of (4-Cyclopropanesulfonylaminocarbonyl-l8-hydroxy-2,15- dioxo-3, 16-diaza-tricyclo[l 4.3.0. G 4 ' 6 ] nonadec- 7-en-14-yl)-carbamic acid tert-butyl ester
  • Step 1 To a solution of 7V-Boc-cysteine methyl ester (3.36 g, 0.014 mol) in methanol (166 mL) at RT was added triethylamine (10.8 mL) and l-bromopent-4-ene (3.19 g, 21 mmol, 1.5 equivalents) and the resulting solution was stirred at room temperature overnight. The mixture was then concentrated in vacuo and the resulting residual mixture was purified using flash chromatography (hexane, ethyl acetate gradient) to provide 1.76 g (41%) of the desired thioether.
  • Step 2 The thioether product of step 1 (9.51 g, 31.4 mmol) was added to a mixture of IM LiOH in water (20OmL) and THF (20OmL) and the resulting mixture was stirred at room temperature overnight.
  • Example 28 Preparation of Example 28, N-tert-Butoxycarbonyl-3-(4-pentenylthio)-L-valine
  • Step 1 Preparation of N-tert-butoxycarbonyl-3-(4-pentenylthio)-L-valine, methyl ester
  • the mixture was concentrated under vacuum, and the residue was dissolved in water.
  • the aqueous mixture was washed with diethyl ether, adjusted to pH 3 employing IN hydrochloric acid, and then extracted with ethyl acetate.
  • the combined extracts were washed with brine, dried over anhydrous magnesium sulfate, filtered, and concentrated under vacuum.
  • the crude product (12.2Og) was dissolved in 12OmL of anhydrous dimethylsulfoxide. To this solution was added 10.50g (76 mmol) of potassium carbonate and 4.7OmL (76 mmol) of iodomethane, and the resulting mixture was stirred at room temperature for 24 hours.
  • reaction mixture was diluted with water and extracted with ethyl acetate.
  • the combined extracts were washed with water (2X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum.
  • Column chromatography on silica gel (elution: 2 -10% ethyl acetate/hexane) provided 8.54g of N-tert-butoxycarbonyl-3-(4-pentenylthio)-L- valine, methyl ester as a colorless oil.
  • Step 3 Preparation of Isopropyl 2(S)-(tert-butoxycarbonylamino)-5- hydroxypentanoate
  • Example 23 was prepared by adding a DMF solution of N-trityl protected threonine to a DMF solution of sodium hydride cooled to — 15 C. The reaction mixture was stirred for 30 minutes at— 15 C after which 5-bromo-l-pentene was added and the resulting mixture was warmed to —5 C. The reaction mixture was maintained at —5 C for 3 days after which time the reaction was quenched by the addition of IN aqueous HCl and worked up using standard extraction procedures as described above. Example 23 was obtained in pure form by standard chromatography procedures.
  • Example 31 Preparation of Example 31, N-tert-Butoxycarbonyl-O-(4-pentenyl)-L-serine
  • the mixture was diluted with 200OmL of water, adjusted to pH 3-4 by the addition of 5OmL of 1.00N hydrochloric acid, and extracted with ethyl acetate.
  • the organic phase was washed with water (2X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum.
  • To remove the residual mineral oil the resulting material was dissolved in a dilute aqueous sodium hydroxide solution. This aqueous solution was washed with hexane and then adjusted to pH 4 employing hydrochloric acid, and extracted with ethyl acetate.
  • the extract was washed with water (2X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum.
  • the crude product (7.7Og) was dissolved in 10OmL of anhydrous dimethylsulfoxide. To this solution was added 7.8Og (56 mmol) of potassium carbonate and 3.5OmL (56 mmol) of iodomethane, and the resulting mixture was stirred at room temperature for 24 hours. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined extracts were washed with water (2X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum.
  • Example 31 To a solution of 6.65g (23 mmol) of N-tert-butoxycarbonyl-0-(4-pentenyl)-L- serine, methyl ester in 50OmL of tetrahydrofuran at room temperature was added a solution of 1.95g (46 mmol) of lithium hydroxide monohydrate in 10OmL of Water. The resulting mixture was stirred at room temperature for 40 hours. To the reaction mixture then was added 46mL of 1.00N hydrochloric acid.
  • Step 1 Preparation ofl4-tert-butoxycarbonylamino-2,15-dioxo-18-(pyridin-2- yloxy)-3, 16-diaza-tricyclo[14.3.0.0 4 ' 6 ]nonadec- 7-ene-4-carboxylic acid
  • the major component was not recovered from the preparative HPLC while the minor component, ( 15,4/?, 65, 145,1 BR)-I -cis- 14-terf-butoxycarbonylamino-l 8-(4-chloroquinolin-8-yloxy)-2,l 5-dioxo-3, 16- diazatricyclo[14.3.0.0 4>6 ]nona-dec-7-ene-4-carboxylic acid, was collected and concentrated into a white foam (1.9 mg, 3%).
  • Step 1 Prepared l-(2(5)-tert-butoxycarbonylamino-non-8-enoyl)-4(i?)- benzyloxy-pyrrolidine-2(-?)-carboxylic acid, methyl ester by way of example 25, step 1 using 5-AUyloxy-2(S)-(tert-butoxycarbonylamino)pentanoic acid (2.77 g, 10.1 mmol; prepared in Ex.
  • Step 2 Prepared l- ⁇ [l-(2(,S)-te ⁇ Butoxycarbonylamino-non-8-enoyl)-4(i?)- benzyloxypyrrolidine-2(S)carbonyl]-(li2)-amino ⁇ -2(S)-vinyl- cyclopropanecarboxylic acid ethyl by way of example 25, step 2 using 1- (2(S)-tert-butoxycarbonylarnino-non-8-enoyl)-4(i?)-benzyloxy-pyrrolidine- 2(S)-carboxylic acid methyl ester (2.78g 5.80 mmol), saponifying and then coupling with (li?,25>l-amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride (0.989 g, 6.38 mmol) to give l- ⁇ [l-(2(S)-tert- Butoxycarbonylamino-non-8-
  • Step 3 Prepared (1S,4R,6S,14S18R, 7-cj,y)-14-tert-butoxycarbonylamino-18- benzyloxy ⁇ lS-dioxo-Sjl ⁇ -diazatricyclotM.S.O.O ⁇ J-nonadec-V-ene ⁇ - carboxylic acid, ethyl ester by way of example 25, step 3 using l- ⁇ [l-(2(5)- tert-butoxycarbonylamino-non-8-enoyl)-4(i?)-benzyloxypyrrolidine- 2(iS)carbonyl]-(li?)-amino ⁇ -2(5)-vinyl-cyclopropanecarboxylic acid ethyl ester (2.71 g, 4.42 mmol) to give (1S,4R,6$,14S, 18R, l-cis)- ⁇ 4-tert- butoxycarbonylamino-18-benzyloxy-2,
  • Step 4 Prepared (1S,4R,6S,14S,18R, 7-cis)-14-tert-butoxycarbonylamino-18- benzyloxy-2,15-dioxo-3,16-diazatricyclo[14.3.0.0 4 ' 6 ]-nonadec-7-ene-4- carboxylic acid by way of example 25, step 4 using (IS,4R, 6S, 14S, 1 SR, 7- cis)- 14-tert-butoxycarbonylamino- 18-benzyloxy-2, 15-dioxo-3, 16- diazatricyclo[14.3.0.0 4 ' 6 ]-nonadec-7-ene-4-carboxylic acid ethyl ester (1.30 g, 2.22 mmol) to give ( ⁇ S,4RfiS, ⁇ 4S, ⁇ %R, 7-cis)-14-tert-butoxycarbonylamino- 18-benzyloxy ⁇ .15-di
  • Step 5 Prepared (1S,4R, 6S, 143, 18R, 7-e/s)-l 8-benzyloxy- 14-tert- butoxycarbonylamino-4-cyclopropanesulfonylaminocarbonyl-2,15-dioxo- 3,16-diaza-10-oxatricyclo[14.3.0.0 4 ' 6 ]nonadec-7-ene by way of example 26, step 4 using (IS,4R,6S,14S, 1 BR, 7-cw)-14-?e ⁇ -butoxycarbonylamino-18- benzyloxy-2,15-dioxo-3,16-diazatricyclo[14.3.0.0 4>6 ]-nonadec-7-ene-4- carboxylic acid (860 mg, 1.51 mmol) and cyclopropylsulfonamide (365 mg, 3.02 mmol) to give (1S,4R,6S,14S, 18R, 7-cz
  • Step 1 A mixture of 3,5-dimethyl-4-nitro-isoxazole (1.42 g, 10.0 mmol), phenylacetaldehyde (1.32 g, 11.0 mmol) in piperidine (1 mL) and ethanol (10 mL) was heated to reflux for 16 h. After cooling down to the ambient temperature, the product precipitated out was collected by filtration. The cake was washed with cold ethanol thoroughly to afford 1.20 g (53%) of the desired product as a white solid.
  • Step 2 A solution of 3-methyl-5-phenyl-isoxazolo[4,5-b]pyridine 4-oxide (1.00 g, 4.40 mmol) and POC13 (2.71 g, 17.7 mmol) in chloroform (10 mL) was heated to reflux for 1 h. After cooling down to the ambient temperature, the final solution was diluted with chloroform (50 mL) and washed with NaHCO3 (aq.) (two 50 mL portions) and brine, dried over MgSO4, filtered, evaporated. The residue was purified by flash chromatography (4:1 hexane-EtOAc) to afford 790 mg (73%) of the desired product as a white solid.
  • Step 1 A mixture of 2-amino-6-methylpyridine (1.08 g, 10.0 mmol), ethyl benzoylacetate (2.30 g, 12.0 mmol) and polyphosphoric acid (6.00 g, 61.2 mmol) was heated to 110 0 C for 5 h. After cooling to the ambient temperature, the mixture was poured into iced water (20 mL) and neutralized to pH 7 with 10 M NaOH. Extracted with CHCI3. The organic layer was washed with brine, dried over MgSO 4 , filtered, evaporated. The residue was purified by flash chromatography (1 : 1 hexane-EtOAc) to afford 510 mg (22%) of the desired product as a pale yellow solid.
  • Step 2 A solution of 6-methyl-2-phenyl-pyrido[l,2a]pyrimidin-4-one (489 mg, 2.07 mmol) in melted diphenyl ether (5 mL) was heated to gentle reflux for 5 h. After cooling to the ambient temperature, the formed suspension was diluted with diethyl ether (10 mL), filtered. The cake was washed with diethyl ether thoroughly to afford 450 mg (92%) of the desired product as a brownish solid.MS m/z 237 (M + +H).
  • Step 3 A suspension of 7-methyl-2-phenyl-lH-[l,8]naphthyridin-4-one (450 mg, 1.91 mmol) in POCl 3 (10 mL) was heated to gentle reflux for 3 h. then evaporated in vacuo. The residue was poured into iced water (20 mL) and neutralized to pH 10 with 10 M NaOH. The mixture was then extracted with CHCl 3 and the organic layer was washed with brine, dried over MgSO 4 , filtered and evaporated. The residue was purified by flash chromatography (2: 1 hexane-EtOAc) to afford 450 mg (92%) of the desired product as a pink solid.
  • Step 1 To a solution of 4-methoxyphenethyl alcohol (1.52 g, 10.0 mmol) in CH2C12 (50 mL) at 0OC was added Dess-Martin reagent (4.45 g, 10.5 mmol) in one portion. The formed mixture was allowed to warm to the ambient temperature for 1 h. Washed with sat. Na2S2O3 (aq) and IM NaOH, brine respectively. Dried over MgSO4, evaporated in vacuo to give 1.50 g (100%) of the desired aldehyde as a viscous oil. This product was used as crude without any further purification.
  • Step 2 A solution of 3,5-dimethyl-4-nitro-isoxazole (142 mg, 1.0 mmol), 4- methoxy-phenylacetaldehyde from Example 3, Step 1 (180 mg, 1.1 mmol) in piperidine (0.1 mL) and ethanol (2 mL) was heated to reflux for 12 h. After cooling down to the ambient temperature, the product precipitated out was collected by filtration. The cake was washed with cold ethanol thoroughly to afford 130 mg (51%) of the desired product as a grayish solid.
  • Step 3 This product was prepared by the same procedure as described in Example 39, Step 2. 1 H NMR (CDCl 3 ) ⁇ 2.70 (s, 3H), 3.87 (s, 3H), 7.00-7.03 (m, 2H), 7.84 (s, IH), 7.96-7.98 (m, 2H); MS m/z 275, 277 (M + +H).
  • Intermediate 41. can be used to make compounds of formula (I) as follows:
  • Interemediate 46 was prepared as described by P. Ferrarini et al, in J Heterocyclic Chem, 1983, plO53. Intermediate 46 can be used to make compounds of formula (I) as follows:
  • Step 2 To a 0 0 C slurry of Boc-c/s-Hydroxyproline-OMe (2.Og, 8.15mmol)and 3 (2.26g, 8.97mmol) in THF (82mL) was added Ph 3 P and diisopropyl azocarboxylate (1.98 g, 8.97mmol). After stirring at rt for 17h, the reaction mixture was diluted with EtOAc (10OmL) and washed with H 2 O (5OmL). The aqueous layer was separated and back-extracted with EtOAc (2 x 5OmL).
  • Stepl Prepared as described in Example 48 and using 2-bromo-4,5- dimethoxybenzoic acid and trifluoroamidine as starting materials.
  • Step 2 As described in Example 48.
  • Step 1 A solution of 3-phenyl-but-2-enoic acid (16.2 g), diphenylphosphoryl azide (27.5 g), and triethylamine (10.1 g) in benzene (100 mL) was stirred for 1 h. After filtration through a silica gel plug washing with benzene and concentration, the residue was dissolved in diphenylmethane (80 mL) and refiuxed for 3 h. After cooling to rt, solids were collected through a plug washing with benzene and dried to give 10 g (63%) of the desired product as a solid.
  • Step 2 A solution of 4-methyl-2H-isoquinolin-l-one (4.8 g) in POCl 3 (50 mL) was refiuxed for 3 h. After cooling and concentration, the residue was based with 5 N NaOH and extracted with CH 2 Cl 2 . The organic layer was washed with brine and dried over MgSO 4 .
  • Step 1 Preparation of 7-fluoro-6-methoxy-2/ ⁇ -isoquinolin-l-one. As shown in step 1 of this example using 19.6 g 4-fluoro-3-methoxycinnamic acid as starting material. 9.5 g product obtained (48% yield).
  • Step 2 Preparation of l-chloro-7-fluoro-6-methoxyisoquinoline: As shown in step 2 of this example using 7-fluoro-6-methoxy-2H-isoquinolin-l-one (9g) as starting material. 7 g of desired product obtained (70% yield).
  • Step 1 As in Example 55 step 1 but with 3.82 g of 3-(4-Fluoro-phenyl)-3-methoxy- acrylic acid as starting material. 198 mg product obtained (5% yield). Product:
  • Step 2 As in Example 55, step 1 , but with 193 mg 7-fluoro-4-methoxy-2H- isoquinolin-1-one as starting material. 199 mg product obtained (94% yield).
  • Step 1 To a solution of Boc-HYP-OH (1.0 g, 4.324 mmol) in DMF (20 mL), NaH (0.38 g of 60% dispersion in mineral oil, 9.513 mmol) was added at 0 0 C. The reaction mixture was stirred for 1 hr. Then 2,4-dichloropyrimidine (0.709 g, 0.0289 mmol) was added. The reaction mixture was warmed to rt and stirred for overnight. It was then quenched with IN HCl solution and extracted with EtOAc. The organic layer was separated, washed with brine and dried (MgSO 4 ). Evaporation of solvent gave crude product which was then purified by Prep. HPLC to give colorless oil as product.
  • Step 2 To a solution of (2S, 4R) 4-(2-Chloro-pyrimidin-4-yloxy)-pyrrolidine-l,2- dicarboxylic acid 1-tert-butyl ester (0.34 g, 0.99 mmol) in CH 3 CN (20 mL) was added (IR, 2S)/(1S, 2R) (l-cyclopropanesulfonyl-aminocarbonyl-2-vinyl-cyclo- propyl)-carbamic acid (0.511 g, 1.48 mmol), DIEA (0.86 mL, 4.95 mmol) and the coupling reagent HOBt (0.226 g, 1.48 mmol) and HBTU (0.561 g, 1.48 mmol).
  • Step 3 To a solution of intermediate 4 (50 mg, 0.061 mmol) in CH 2 Cl 2 (2.5 mL), 1,2,3,4-tetrahydroisoquinoline (0.011 mL, 0.0915 mmol) and Et 3 N (0.021 mL, 0.153 mmol) were added.
  • Step 4 To a solution of 4-[2-(3,4-Dihydro-lH-isoquinolin-2-yl)-pyrimidin-4-yloxy]- pyrrolidine-2-carboxylic acid (l-cyclopropanesulfonylaminocarbonyl-2-vinyl- cyclopropyl)-amide hydrochloride (20 mg, 0.032 mmol) in CH 3 CN (5 mL) was added 2-methoxycarbonylamino-3,3-dimethyl-butyric acid (9.1 mg, 0.048 mmol), DIEA (0.028 mL, 0.16 mmol) and the coupling reagent HOBt (7.3 mg, 0.048 mmol) and HBTU (18.2 mg, 0.048 mmol).
  • Step 2 To a solution of the proline carboxylic acid (270 mg, 1.1 mmol) in DMSO (10 mL), potassium t-butoxide (309 mg, 2.75 mmol) was added. The reaction mixture was stirred at rt for lhr. Then 2-Bromo-4-chloro-pyridine (254 mg, 1.32 mmol) was added. The reaction mixture was stirred at rt for overnight.
  • Example 68-72 the intermediates described (68-72) and the proposed compounds of formula (I) 5 each contain a halopyridine functionality. This functionality can be employed in coupling reactions wherein the halo group is replace with a ring system or alternate functionality. This reaction is well recognized in the art and the following reactions serve as examples of said coupling process.
  • Example 73 Coupling Reaction: Example A
  • Step 1 The tosylate of the Boc proline intermediate (A) was prepared as described in the literature (Patchett, A. A.; Witkof, B. J. Am. Chem. Soc. 1957, 185-192) and was used without further purification.
  • Step 1 To a solution of commercially available N-Boc-(4S)-(cis)-Hydroxyproline- OMe (200mgs, 0.82 mmole), triphenylphosphine (320mgs, 1.22 mmole) and 1- naphthol (176mgs, 1.22 mmole) in 2.5 mL tetrahydrofuran was added dropwise a solution of diethyldiazodicarboxylate (190 ⁇ L, 1.22 mmole) in 1.0 mL THF over 10 minutes. After stirring for 5.5 days, the reaction was concentrated in vacuo.
  • the crude yellow oil was chromatographed on a 20X40cM preparative TLC plate (Analtech SiO2) eluting with 6-1 hexanes-ethyl acetate to yield the desired product as a pale yellow oil (150mgs, 33%).
  • Step 3 To a solution of Boc-((4R)-naphthal-l-oxo)-Pro-OH (147mgs, 0.41 mmole) and racemic (lR/2S)/(lS/2R)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride salt (79mgs, 0.41 mmole) in 2.8mL methylene chloride was added DIPEA (250 ⁇ L, 1.44 mmole) and TBTU (158mgs, 0.49 mmole). The resulting solution was stirred under nitrogen for 20 hours and then diluted with 4OmL methylene chloride.
  • Diastereomer B Example 1OB: P2[Boc(4R)-(naphthal-l-oxo)proline]-Pl(lS, 2R
  • HCV NS3/4A protease complex enzyme assays and cell-based HCV replicon assays were utilized in the present disclosure, and were prepared, conducted and validated as follows: Generation of recombinant HCV NS3/4A protease complex
  • HCV NS3 protease complexes derived from the BMS strain, H77 strain or J4L6S strain, were generated, as described below. These purified recombinant proteins were generated for use in a homogeneous assay (see below) to provide an indication of how effective compounds of the present disclosure would be in inhibiting HCV NS3 proteolytic activity.
  • Serum from an HCV-infected patient was obtained from Dr. T. Wright, San Francisco Hospital.
  • An engineered full-length cDNA Ccompliment deoxyribonucleic acid) template of the HCV genome CBMS strain was constructed from DNA fragments obtained by reverse transcription-PCR CRT-PCR) of serum RNA Cribonucleic acid) and using primers selected on the basis of homology between other genotype Ia strains. From the determination of the entire genome sequence, a genotype Ia was assigned to the HCV isolate according to the classification of Simmonds et al. (See P Simmonds, KA Rose, S Graham, SW Chan, F McOmish, BC Dow, EA Follett, PL Yap and H Marsden, J. Clin.
  • the amino acid sequence of the nonstructural region, NS2-5B was shown to be >97% identical to HCV genotype Ia (H77) and 87% identical to genotype Ib (J4L6S).
  • the infectious clones, H77 (Ia genotype) and J4L6S (Ib genotype) were obtained from R. Purcell (NIH) and the sequences are published in Genbank (AAB67036, see Yanagi, M., Purcell, R.H., Emerson, S.U. and Bukh, J. Proc. Natl. Acad. ScL U.S.A.
  • the H77 and J4L6S strains were used for production of recombinant NS 3/4 A protease complexes.
  • DNA encoding the recombinant HCV NS3/4A protease complex (amino acids 1027 to 1711) for these strains were manipulated as described by P. Gallinari et al. (see Gallinari P, Paolini C, Brennan D, Nardi C, Steinkuhler C, De Francesco R. Biochemistry. 38(17):5620-32, (1999)). Briefly, a three-lysine solubilizing tail was added at the 3'-end of the NS4A coding region.
  • the cysteine in the Pl position of the NS4A-NS4B cleavage site was changed to a glycine to avoid the proteolytic cleavage of the lysine tag. Furthermore, a cysteine to serine mutation was introduced by PCR at amino acid position 1454 to prevent the autolytic cleavage in the NS 3 helicase domain.
  • the variant DNA fragment was cloned in the pET21b bacterial expression vector (Novagen) and the NS3/4A complex was expressed in Escherichia, coli strain BL21 (DE3) (Invitrogen) following the protocol described by P. Gallinari et al.
  • the cells were resuspended in lysis buffer (10 mL/g) consisting of 25 mM iV-(2-Hydroxyethyl)Piperazine-iV l -(2-Ethane Sulfonic acid) (HEPES), pH 7.5, 20% glycerol, 500 mM Sodium Chloride (NaCl), 0.5% Triton X-100, 1 microgram/milliliter (“ ⁇ g/mL”) lysozyme, 5 mM Magnesium Chloride (MgCl 2 ), 1 ⁇ g/ml Dnasel, 5mM ⁇ -Mercaptoethanol ( ⁇ ME), Protease inhibitor-Ethylenediamine Tetraacetic acid (EDTA) free (Roche), homogenized and incubated for 20 minutes (min) at 4°C.
  • lysis buffer 10 mL/g
  • ⁇ ME ⁇ g/ml
  • ⁇ ME Protease inhibitor-Eth
  • the homogenate was sonicated and clarified by ultra-centrifugation at 235000 g for 1 h at 4°C. Imidazole was added to the supernatant to a final concentration of 15 mM and the pH adjusted to 8.0.
  • the crude protein extract was loaded on a Nickel-Nitrilotriacetic acid (Ni-NTA) column pre- equilibrated with buffer B (25 mM HEPES, pH 8.0, 20% glycerol, 500 mM NaCl, 0.5% Triton X-IOO 5 15 mM imidazole, 5 mM ⁇ ME). The sample was loaded at a flow rate of 1 mL/min.
  • the column was washed with 15 column volumes of buffer C (same as buffer B except with 0.2% Triton X-100).
  • the protein was eluted with 5 column volumes of buffer D (same as buffer C except with 200 mM Imidazole).
  • NS3/4A protease complex-containing fractions were pooled and loaded on a desalting column Superdex-S200 pre-equilibrated with buffer D (25 mM HEPES, pH 7.5, 20% glycerol, 300 mM NaCl, 0.2% Triton X-100, 10 mM ⁇ ME). Sample was loaded at a flow rate of 1 mL/min. NS3/4A protease complex-containing fractions were pooled and concentrated to approximately 0.5 mg/ml. The purity of the
  • NS3/4A protease complexes derived from the BMS, H77 and J4L6S strains, were judged to be greater than 90% by SDS-PAGE and mass spectrometry analyses.
  • the enzyme was stored at -80 0 C, thawed on ice and diluted prior to use in assay buffer.
  • FRET peptide assay to monitor HCV NS3/4A proteolytic activty The purpose of this in vitro assay was to measure the inhibition of HCV NS3 protease complexes, derived from the BMS strain, H77 strain or J4L6S strain, as described above, by compounds of the present disclosure.
  • This assay provides an indication of how effective compounds of the present disclosure would be in inhibiting HCV NS 3 proteolytic activity.
  • an NS3/4A peptide substrate was used.
  • the substrate was RET Sl (Resonance Energy Transfer Depsipeptide Substrate; AnaSpec, Inc. cat # 2299I)(FRET peptide), described by Taliani et al. in Anal. Biochem. 240(2):60-67 (1996).
  • the sequence of this peptide is loosely based on the NS4A/NS4B natural cleavage site for the HCV NS3 protease except there is an ester linkage rather than an amide bond at the cleavage site.
  • the peptide also contains a fluorescence donor, EDANS, near one end of the peptide and an acceptor, DABCYL, near the other end.
  • EDANS fluorescence donor
  • DABCYL acceptor
  • the fluorescence of the peptide is quenched by intermolecular resonance energy transfer (RET) between the donor and the acceptor, but as the NS3 protease cleaves the peptide the products are released from RET quenching and the fluorescence of the donor becomes apparent.
  • RET intermolecular resonance energy transfer
  • the peptide substrate was incubated with one of the three recombinant NS3/4A protease complexes, in the absence or presence of a compound of the present disclosure.
  • the inhibitory effects of a compound was determined by monitoring the formation of fluorescent reaction product in real time using a Cytofluor Series 4000.
  • the reagents were as follow: HEPES and Glycerol (Ultrapure) were obtained from GIBCO-BRL.
  • Dimethyl Sulfoxide (DMSO) was obtained from Sigma, ⁇ - Mercaptoethanol was obtained from Bio Rad.
  • Assay buffer 50 mM HEPES, pH 7.5; 0.15 MNaCl; 0.1% Triton; 15% Glycerol; 10 mM ⁇ ME.
  • Substrate 2 ⁇ M final concentration (from a 2 mM stock solution in DMSO stored at -20 0 C).
  • HCV NS3/4A protease type Ia (Ib) 2-3 nM final concentration (from a 5 ⁇ M stock solution in 25 mM HEPES, pH 7.5, 20% glycerol, 300 mM NaCl, 0.2% Triton-XIOO, 10 mM ⁇ ME).
  • the assay was made more sensitive by adding 50 ⁇ g/ml Bovine Serum Albumin (Sigma) to the assay buffer and reducing the end protease concentration to 300 pM.
  • the assay was performed in a 96-well polystyrene black plate from Falcon. Each well contained 25 ⁇ l NS3/4A protease complex in assay buffer, 50 ⁇ l of a compound of the present disclosure in 10% DMSO/assay buffer and 25 ⁇ l substrate in assay buffer. A control (no compound) was also prepared on the same assay plate. The enzyme complex was mixed with compound or control solution for 1 min before initiating the enzymatic reaction by the addition of substrate. The assay plate was read immediately using the Cytofluor Series 4000 (Perspective Biosystems). The instrument was set to read an emission of 340 nm and excitation of 490 nm at 25°C. Reactions were generally followed for approximately 15 min.
  • the specificity assays were performed to demonstrate the in vitro selectivity of the compounds of the present disclosure in inhibiting HCV NS3/4A protease complex as compared to other serine or cysteine proteases.
  • Each pNA assay included a 2 h enzyme-inhibitor pre-incubation at room temperature followed by addition of substrate and hydrolysis to ⁇ 15% conversion as measured on a Spectramax Pro microplate reader.
  • the cathepsin B assay was initiated by adding substrate to a 10 min enzyme-inhibitor pre-incubation at room temperature, and the assay plate measured immediately using the Cytofluor Series 4000. Compound concentrations varied from 100 to 0.4 ⁇ M depending on their potency.
  • Tris(hydroxymethyl)aminomethane hydrochloride pH 8
  • Tris-HCl pH 8
  • 0.5 M Sodium Sulfate Na 2 SO 4
  • 50 mM NaCl 50 mM NaCl
  • 0.1 mM EDTA 50 mM NaCl
  • 0.1 mM EDTA 50 mM NaCl
  • 0.1 mM EDTA 50 mM NaCl
  • 0.1 mM EDTA 3% DMSO
  • Tween- 20 with: 133 ⁇ M succ-AAA-pNA and 20 nM HNE or 8 nM PPE; 100 ⁇ M succ-AAPF-pNA and 250 pM Chymotrypsin.
  • the percentage of inhibition was calculated using the formula: [ 1 -((UV inh -UV blank )/(UV cll -UV blank ))] x 100
  • the replicon consists of (i) the HCV 5' UTR fused to the first 12 amino acids of the capsid protein, (ii) the neomycin phosphotransferase gene (neo), (iii) the IRES from encephalomyocarditis virus (EMCV), and (iv) HCV NS3 to NS5B genes and the HCV 3 ' UTR. Plasmid DNAs were linearized with Seal and RNA transcripts were synthesized in vitro using the T7 MegaScript transcription kit (Ambion, Austin, TX) according to manufacturer's directions. In vitro transcripts of the cDNA were transfected into the human hepatoma cell line, HUH-7. Selection for cells constitutively expressing the HCV replicon was achieved in the presence of the selectable marker, neomycin (G418). Resulting cell lines were characterized for positive and negative strand RNA production and protein production over time.
  • the HCV replicon FRET assay was developed to monitor the inhibitory effects of compounds described in the disclosure on HCV viral replication.
  • HUH-7 cells constitutively expressing the HCV replicon, were grown in Dulbecco's Modified Eagle Media (DMEM) (Gibco-BRL) containing 10% Fetal calf serum (FCS) (Sigma) and 1 mg/ml G418 (Gibco-BRL). Cells were seeded the night before (1.5 x 10 4 cells/well) in 96-well tissue-culture sterile plates.
  • DMEM Dulbecco's Modified Eagle Media
  • FCS Fetal calf serum
  • G418 G418
  • Compound and no compound controls were prepared in DMEM containing 4% FCS 5 1 : 100 Penicillin/Streptomysin (Gibco-BRL), 1:100 L-glutamine and 5% DMSO in the dilution plate (0.5% DMSO final concentration in the assay).
  • Compound/DMSO mixes were added to the cells and incubated for 4 days at 37°C. After 4 days, cells were first assessed for cytotoxicity using alamar Blue (Trek Diagnotstic Systems) for a CC50 reading. The toxicity of compound (CC50) was determined by adding 1/10 th volume of alamar Blue to the media incubating the cells.
  • the cells were lysed with 25 ⁇ l of a lysis assay reagent containing an HCV protease substrate (5X cell Luciferase cell culture lysis reagent (Promega #E153A) diluted to IX with distilled water, NaCl added to 150 mM final, the FRET peptide substrate (as described for the enzyme assay above) diluted to 10 ⁇ M final from a 2 mM stock in 100% DMSO.
  • the HCV protease substrate The plate was then placed into the Cytofluor 4000 instrument which had been set to 340 nm excitation/490 nm emission, automatic mode for 21 cycles and the plate read in a kinetic mode. EC 50 determinations were carried out as described for the IC50 determinations.
  • EC50 determinations from the replicon FRET assay were confirmed in a replicon luciferase reporter assay.
  • Utilization of a replicon luciferase reporter assay was first described by Krieger et al (Krieger N, Lohmann V, and Bartenschlager R, J. Virol. 75(10):4614-4624 (2001)).
  • the replicon construct described for our FRET assay was modified by inserting cDNA encoding a humanized form of the Renilla luciferase gene and a linker sequence fused directly to the 3 '-end of the luciferase gene.
  • This insert was introduced into the replicon construct using an Ascl restriction site located in core, directly upstream of the neomycin marker gene.
  • the adaptive mutation at position 1179 (serine to isoleucine) was also introduced (Blight KJ, Kolykhalov, AA, Rice, CM, Science 290(5498): 1972-1974).
  • a stable cell line constitutively expressing this HCV replicon construct was generated as described above.
  • the luciferase reporter assay was set up as described for the HCV replicon FRET assay with the following modifications. Following 4 days in a 37°C/5% CO 2 incubator, cells were analyzed for Renilla Luciferase activity using the Promega Dual-Glo Luciferase Assay System.
  • % control average luciferase signal in experimental wells (+ compound) average luciferase signal in DMSO control wells (- compound)
  • % control average luciferase signal in experimental wells (+ compound) average luciferase signal in DMSO control wells (- compound)
  • % control average luciferase signal in experimental wells (+ compound) average luciferase signal in DMSO control wells (- compound)
  • % control average luciferase signal in experimental wells (+ compound) average luciferase signal in DMSO control wells (- compound)
  • the values were graphed and analyzed using XLfit to obtain the EC50 value.
  • Representative compounds of the disclosure were assessed in the HCV enzyme assays, HCV replicon cell assay and/or in several of the outlined specificity assays.
  • Compound 3 was found to have an IC50 of 2.8 nanomolar (nM) against the NS3/4A BMS strain in
  • IC 50 Activity Ranges (NS3/4A BMS Strain): A is > 1 micromolar ( ⁇ M); B is 0.1-1 ⁇ M; C is ⁇ 0.1 ⁇ M EC50 Activity Range (for compounds tested): A is > 1 ⁇ M; B is 0.1-1 ⁇ M; C is ⁇ 0.1 ⁇ M
  • the compounds have a biological activity (EC 50 ) of 100 ⁇ M or less, and in another embodiment, 1 ⁇ M or less, and most preferably 0.1 ⁇ M or less.
  • Table 3 is a list of compounds that could be synthesized using the teachings described or referenced herein.

Abstract

Macrocyclic peptides are disclosed having the general formula: [I] , wherein R', R3, R3', R4, R6, X5 Q, and W are described. Compositions comprising the compounds and methods for using the compounds to inhibit HCV are also disclosed.

Description

HEPATITIS C VIRUS INHIBITORS
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application Serial
Numbers 60/736,606 filed November 14, 2005 and 60/733,039 filed November 3, 2005.
The present disclosure is generally directed to antiviral compounds, and more specifically directed to compounds which inhibit the functioning of the NS 3 protease (also referred to herein as "serine protease") encoded by Hepatitis C virus (HCV)3 compositions comprising such compounds and methods for inhibiting the functioning of the NS3 protease.
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. (Lauer, G. M.; Walker, B. D. N. Engl. J. Med. (2001), 345, 41-52).
Presently, the most effective HCV therapy employs a combination of alpha- interferon and ribavirin, leading to sustained efficacy in 40% of patients. (Poynard, T. et al. Lancet (1998), 352, 1426-1432). Recent clinical results demonstrate that pegylated alpha-interferon is superior to unmodified alpha-interferon as monotherapy (Zeuzem, S. et al. N. Engl. J. Med. (2000), 343, 1666-1672). However, even with experimental therapeutic regimens involving combinations of pegylated alpha- interferon and ribavirin, a substantial fraction of patients do not have a sustained reduction in viral load. Thus, there is a clear and long-felt need to develop effective therapeutics for treatment of HCV infection.
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. Considerable heterogeneity is found within the nucleotide and encoded amino acid sequence throughout the HCV genome. At least six major genotypes have been characterized, and more than 50 subtypes have been described. The major genotypes of HCV differ in their distribution worldwide, and the clinical significance of the genetic heterogeneity of HCV remains elusive despite numerous studies of the possible effect of genotypes on pathogenesis and therapy.
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. The first one is believed to cleave at the NS2-NS3 junction; the second one is a serine protease contained within the N-terminal region of NS3 and mediates all the subsequent cleavages downstream of NS3, both in cis, at the NS3-NS4A cleavage site, and in trans, for the remaining NS4A- NS4B, NS4B-NS5A, NS5 A-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. The complex formation of the NS3 protein with NS4A seems necessary to the processing events, enhancing the proteolytic efficiency at all of the sites. The NS3 protein also exhibits nucleoside triphosphatase and RNA helicase activities. NS5B is a RNA-dependent RNA polymerase that is involved in the replication of HCV.
1. In a first aspect the present disclosure provides A compound of formula (I)
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein
(a) R »4 is H; Ci-6 alkyl, or C3-7 cycloalkyl each optionally substituted with halo; alkoxy; -C(O)-R5; C(O)-N(R5)2; or C(O)-OR5; wherein each R5 is independently Ci-9 alkyl, optionally substituted with C1-6 alkoxy, C3-7 cycloalkoxy, halo-Ci-6 alkoxy, cyano, halo, hydroxy, amino, Ci-6 alkylamino, di (Ci-6) alkylamino, di (Ci-6) alkylamide, carboxyl, (Ci -β) carboxyester; Ce-io aryl; C7-14 alkylaryl; heterocylclyl; or C3-7 cycloalkyl optionally substituted with alkoxy, halo, or haloalkoxy;
(b) R6 is H, C1-6 alkyl, or C3-7 cycloalkyl;
(c) R3 and R3' are each independently H or methyl; (d) Q is a C3.9 saturated or unsaturated chain, optionally containing one to three heteroatoms independently selected from O or S(O)m; wherein m is 0, 1 or 2;
(e) W is OH, -O-R1, or -NH-SO2-R2; wherein R1 is d.6 alkyl, unsaturated C3-7 cycloalkyl, C6-I4 aryl or C7-16 alkylaryl; and R2 is Ci-8 alkyl, C4-I0 alkylcycloalkyl, unsubstituted C3-7 cycloalkyl; or R2 is cyclopropyl or cyclobutyl optionally substituted with C1^ alkyl, C2-5 alkenyl, C7.16 alkylaryl, alkoxy, alkoxyalkyl, Cs-7 cycloalkyl, Cs_7 cycloalkenyl, Cβ-\ 0 alkylcycloalkyl, halo, haloalkyl, cyano, alkylcyano, haloalkoxy, or C(O)-X; wherein the C5-7 cycloalkyl, the Cs-7 cycloalkenyl, and the C6-1O alkylcycloalkyl are further optionally substituted with C1^ alkyl or hydroxy; and wherein X is selected from phenyl and -NHRX; wherein Rx is selected from C1-6 alkyl, Het, and Cβ-io aryl;
(f) X is O, S, SO, SO2, OCH2, CH2O or NH;
(g) R' is Het, C6-Io aryl or C7-I4 alkylaryl, each optionally substituted with one or more Ra groups; or R' is C3-9 cycloalkyl or Ci-7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; provided that when X is O, then R' is other than
Figure imgf000004_0001
wherein Ra> is H or Ra; Rx is selected from H, halo, Ci_6 alkyl, Ci-6 alkoxy, C3-6 cycloalkyloxy, aryl, and Het; and Ry is selected from H, halo, CF3, Ci-6 alkoxy, and C3-6 cycloalkyloxy; and (h) each Ra is Ci-β alkyl, C3-7 cycloalkyl, CL6 alkoxy, C3-7 cycloalkoxy, halo-Ci-6 alkyl, CF3, mono-or di-halo-Ci.6 alkoxy, cyano, halo, thioalkyl, hydroxy, alkanoyl, NO2, SH, amino, Ci-β alkylamino, di (Cue) alkylamino, di (d-e) alkylamide, carboxyl, (Ci-6) carboxyester, Ci-6 alkylsulfone, Ci-βalkylsulfonamide, di (Cue) alkyl(alkoxy)amine, C6-Io aryl> C7-14 alkylaryl, or a 5-7 membered monocyclic heterocycle.
In a first embodiment of the first aspect the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein X is O.
In a second embodiment of the first aspect the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein X is O and R' is other than quinoline or isoquinoline.
In a third embodiment of the first aspect the present disclosure provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein X is selected from S, SO, SO2, OCH2, CH2O and NH. In a second aspect the present disclosure provides a compound of formula (II)
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof, wherein
(a) R4 is -C(O)-R5, C(O)-NHR5 or C(O)-OR5; wherein R5 is Ci-9 alkyl optionally substituted with C1.6 alkoxy, cyano, halo;
(b) Q is a C5-7 saturated or unsaturated chain optionally containing one or two heteroatoms independently selected from O and S(O)m; wherein m is 0, 1 or 2;
(c) W is OH or NH-SO2-R2, wherein R2 is Ci-8 alkyl, C4-I0 alkylcycloalkyl, unsubstituted C3.7 cycloalkyl, or cyclopropyl or cyclobutyl optionally substituted with Ci-4 alkyl or C7-Ig alkylaryl;
(d) X is O, S, OCH2, CH2O or NH; (e) R' is Het, C6-I0 aryl or C7-M alkylaryl, each optionally substituted with one or more Ra; or R' is C3.9 cycloalkyl or Ci-7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; provided that when X is O, then R' is other than
Figure imgf000006_0001
.wherein Ra' is H or Ra; Rx is selected from H, halo, C1 -β alkyl, Cj-6 alkoxy, C3-6 cycloalkyloxy, aryl, and Het; and Ry is selected from H, halo, CF3, C1-6 alkoxy, and C3-6 cycloalkyloxy; and
(f) each Ra is C i-6 alkyl, C3-7 cycloalkyl, Ci-6 alkoxy, C3.7 cycloalkoxy, halo-Cj-.β alkyl, CF3, ImIo-Ci-6 alkoxy, cyano, halo, thioalkyl, hydroxy , amino, Ci-βalkylamino, di (Ci-β) alkylamino, di (Ci-β) alkylamide, carboxyl, (Q-Θ) carboxyester, Cι-6 alkylsulfone, Ci-β alkylsulfonamide, di (Ci-6) alkyl(alkoxy)amine, Cβ-io aryl, C7-H alkylaryl, or a 5-7 membered monocyclic heterocycle.
In a first embodiment of the second aspect the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein X is O.
In a second embodiment of the second aspect the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein X is O and R' is other than quinoline or isoquinoline.
In a third embodiment of the second aspect the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein R4 is C(O)-OR5; wherein R5 is Ci-9 alkyl.
In a fourth embodiment of the second aspect the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein Q is a C5./7 saturated or unsaturated chain optionally containing one oxygen atom. In a fifth embodiment of the second aspect the present disclosure provides a compound of formula (II), or a pharmaceutically acceptable salt thereof, wherein W is NH-SO2-R2; wherein R2 is unsubstituted C3_7 cycloalkyl.
In a third aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
In a first embodiment of the third aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier.
In a second embodiment of the third aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier, wherein at least one of the additional compounds is an interferon or ribavirin.
In a third embodiment of the third aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier, wherein at least one of the additional compounds is an interferon or ribavirin, wherein the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastiod interferon tau.
In a fourth embodiment of the third aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier, wherein at least one of the additional compounds is selected from interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti- sense RNA, Imiqimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
In a fifth embodiment of the third aspect the present disclosure provides a composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, one or two additional compounds having anti-HCV activity, and a pharmaceutically acceptable carrier, wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH. In a fourth aspect the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof. In a first embodiment of the fourth aspect the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof and administering one or two additional compounds prior to, after, or simultaneously with the compound of formula (I), or the therapeutically acceptable salt thereof. In a second embodiment of the fourth aspect at least one of the additional compounds is an interferon or ribavirin. In a third embodiment of the fourth aspect the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A5 lymphoblastiod interferon tau. In a fourth embodiment of the fourth aspect the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof and administering one or two additional compounds prior to, after, or simultaneously with the compound of formula (I), or the therapeutically acceptable salt thereof, wherein at least one of the additional compounds is selected from interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'-monophosρate dehydrogenase inhibitor, amantadine, and rimantadine. In a fifth embodiment of the fourth aspect the present disclosure provides a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof and administering one or two additional compounds prior to, after, or simultaneously with the compound of formula (I), or the therapeutically acceptable salt thereof, wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH. In addition, the present disclosure provides macrocyclic isoquinoline compounds of the following formula:
Figure imgf000009_0001
wherein:
(a) R4 is H;
Figure imgf000009_0002
alkyl, or C3-7 cycloalkyl each optionally substituted with halo; alkoxy; -C(O)-R5; C(O)-NHR5; or C(O)-OR5; wherein R5 is C 1.9 alkyl, optionally substituted with C i_6 alkoxy, C3-7 cycloalkoxy, halo-Ci-6 alkoxy, cyano, halo, hydroxy, amino, Ci-βalkylamino, di (Q-e) alkylamino, di (Cj-e) alkylamide, carboxyl, (Ci-g) carboxyester; C6-10 aryl; C7-H alkylaryl; heterocylclyl; or C3-7 cycloalkyl;
(b) Re is H, Ci-6 alkyl, or C3-7 cycloalkyl;
(c) R3 and R'3 are each independently H or methyl;
(d) Q is a C3.9 saturated or unsaturated chain, optionally containing one to three heteroatoms independently selected from O or S(O)n,; wherein m is 0, 1 or 2; (e) W is OH, -O-Rj, or -NH-SO2-R2: wherein Ri is Ci-6 alkyl, unsaturated C3-7 cycloalkyl, C6-I4 aryl or C7-I6 alkylaryl; and R2 is Ci-8 alkyl, C4.10 alkylcycloalkyl, unsubstituted C3-7 cycloalkyl; or R2 is cyclopropyl or cyclobutyl optionally substituted with C1^ alkyl or C7. i6 alkylaryl, alkoxy, halo, haloalkyl, cyano, alkylcyano, or haloalkoxy; (f) X is O, S, SO, SO2, OCH2, CH2O or NH;
(g) R! is Het, Ce- 10 aryl or C7-W alkylaryl, each optionally substituted with Ra; or C3-9 cycloalkyl or Ci-7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; and (h) Ra is Ci-6 alkyl, C3-7 cycloalkyl, Ci-6 alkoxy, C3-7 cycloalkoxy, halo-Ci-.6 alkyl, CF3, mono-or di- halo-C|.6 alkoxy, cyano, halo, thioalkyl, hydroxy, alkanoyl, NO2, SH, , amino, Ci-βalkylamino, di (d-β) alkylamino, di (Cr6) alkylamide, carboxyl, (Ci-β) carboxyester, Ci-βalkylsulfone, Ci-ealkylsulfonamide, di (Ci-g) alkyl(alkoxy)amine, C6-Io aryl, C7-I4 alkylaryl, or a 5-7 membered monocyclic heterocycle. The present disclosure also provides compositions comprising the compounds or pharmaceutically acceptable salts, solvates or prodrugs thereof and a pharmaceutically acceptable carrier. In particular, the present disclosure provides pharmaceutical compositions useful for inhibiting HCV NS3 protease comprising a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier.
The present disclosure further provides methods for treating patients infected with HCV, comprising administering to the patient a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt, solvate or prodrug thereof. Additionally, the present disclosure provides methods of inhibiting HCV NS3 protease by contacting the NS3 protease with a compound of the present disclosure.
By virute of the'present disclosure, it is now possible to provide improved drugs comprising the compounds of the disclosure which can be effective in the treatment of patients infected with HCV. Specifically, the present disclosure provides peptide compounds that can inhibit the functioning of theMS3 protease, e.g., in combination with the NS4A protease. Further, the present disclosure makes it possible to administer combination therapy to a patient whereby a compound in accordance with the present disclosure, which is effective to inhibit the HCV NS3 protease, can be administered with another compound having anti-HCV activity, e.g., a compound which is effective to inhibit the function of a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH and a nucleoside analog for the treatment of an HCV infection.
Stereochemical definitions and conventions used herein generally follow McGraw-Hill Dictionary of Chemical Terms, S. P. Parker, Ed., McGraw-Hill Book Company, New York (1984) and Stereochemistry of Organic Compounds, Eliel, E. and Wilen, S., John Wiley & Sons, Inc., New York (1994). Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, 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 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory and (+) or d, meaning the compound, is dextrorotatory. For a given chemical structure, these compounds, called stereoisomers, are identical except that they are mirror images of one another. A specific stereoisomer of a mirror image pair may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. With reference to the instances where (R) or (S) is used, it is to designate the absolute configuration of a substituent in context to the whole compound and not in context to the substituent alone. Unless otherwise specifically noted herein, the terms set forth below will have the following definitions.
The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
The term "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.
The term "stereoisomers" refers to compounds which have identical chemical composition, but differ with regard to the arrangement of the atoms or groups in space. The term "diastereomer" refers to a stereoisomer which is not an enantiomer, e.g., 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, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography.
The term "enantiomers" refers to two stereoisomers of a compound which are non-superimposable mirror images of one another. The term "pharmaceutically acceptable salt" is intended to include nontoxic salts synthesized from a compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, PA, 1990, p. 1445. The compounds of the present disclosure are useful in the form of the free base or acid or in the form of a pharmaceutically acceptable salt thereof. All forms are within the scope of the disclosure.
The term "therapeutically effective amount" means the total amount of each active component that is sufficient to show a meaningful patient benefit, e.g., a sustained reduction in viral load. When applied to an individual active ingredient, administered alone, the term refers to that ingredient alone. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.
The term "compounds of the disclosure", and equivalent expressions, are meant to embrace compounds of formula (I), and pharmaceutically acceptable enantiomer, diastereomer salts, and solvates, e.g. hydrates, and prodrugs. Similarly, references to intermediates, are meant to embrace their salts, and solvates, where the context so permits. References to the compound of the disclosure also include the preferred compounds, e.g. formula (II) and A-M. The term "derivative" means a chemically modified compound wherein the modification is considered routine by the ordinary skilled chemist, such as an ester or an amide of an acid, protecting groups, such as a benzyl group for an alcohol or thiol, and tert-butoxycarbonyl group for an amine.
The term "solvate" means a physical association of a compound of this disclosure with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolable solvates. Exemplary solvates include hydrates, ethanolates, methanolates, isopropanolates and the like.
The term "prodrug" as used herein means derivatives of the compounds of the disclosure which have chemically or metabolically cleavable groups and become, by solvolysis or under physiological conditions, the compounds of the disclosure which are pharmaceutically active in vivo. A prodrug of a compound may be formed in a conventional manner with a functional group of the compounds such as with an amino, hydroxy or carboxy group when present. The prodrug derivative form often offers advantages of solubility, tissue compatibility, or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985). Prodrugs include acid derivatives well known to practitioners of the art, such as, for example, esters prepared by reaction of the parent acidic compound with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a suitable amine. The term "patient" includes both human and other mammals.
The term "pharmaceutical composition" means a composition comprising a compound of the disclosure in combination with at least one additional pharmaceutical carrier, i.e., adjuvant, excipient or vehicle, such as diluents, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, , perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms. Ingredients listed in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, PA (1999) for example, may be used. The phrase "pharmaceutically acceptable" is employed herein to refer 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 risk/benefit ratio. The term "treating" refers to: (i) preventing a disease, disorder or condition from occurring in a patient which may be predisposed to the disease, disorder and/or condition but has not yet been diagnosed as having it; (ii) inhibiting the disease, disorder or condition, i.e., arresting its development; and (iii) relieving the disease, disorder or condition, i.e., causing regression of the disease, disorder and/or condition.
The term "substituted" as used herein includes substitution at from one to the maximum number of possible binding sites on the core, e.p., organic radical, to which the subsitutent is bonded, e.g., mono-, di-, tri- or tetra- substituted, unless otherwise specifically stated.
The nomenclature used to describe organic radicals, e.g., hydrocarbons and substituted hydrocarbons, generally follows standard nomenclature known in the art, unless otherwise specifically defined. Combinations of groups, e.g., alkylalkoxyamine or arylalkyl, include all possible stable configurations, unless otherwise specifically stated. Certain radicals and combinations are defined below for purposes of illustration.
The term "halo" as used herein means a halogen substituent selected from bromo, chloro, fluoro or iodo. The term "haloalkyl" means an alkyl group that in substituted with one or more halo substituents.
The term "alkyl" as used herein means acyclic, straight or branched chain alkyl substituents having the specified number of carbon atoms and includes, for example, methyl, ethyl, propyl, butyl, tert-butyl, hexyl, 1-methylethyl, 1- methylpropyl, 2-methypropyl, 1,1-dimethylethyl. Thus, Ci-e alkyl refers to an alkyl group having from one to six carbon atoms. The term "lower alkyl" means an alkyl group having from one to six, preferably from one to four carbon atoms. The term "alkylester" means an alkyl group additionally containing on ester group. Generally, a stated carbon number range, e.g., C2-6 alkylester, includes all of the carbon atoms in the radical. The term "alkenyl" as used herein means an alkyl radical containing at least one double bond, e.g., ethenyl (vinyl) and alkyl.
The term "alkoxy" as used herein means an alkyl group with the indicated number of carbon atoms attached to an oxygen atom. Alkoxy includes, for example, methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy and 1,1-dimethylethoxy. The latter radical is referred to in the art as tert-butoxy. The term "alkoxycarbonyl" means an alkoxy group additionally containing a carbonyl group.
The term "cycloalkyl" as used herein means a cycloalkyl substituent containing the indicated number of carbon atoms and includes, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and spiro cyclic groups such as spirocyclopropyl as spirocyclobutyl. The term "cycloalkoxy" as used herein means a cycloalkyl group linked to an oxygen atom, such as, for example, cyclobutyloxy or cyclopropyloxy. The term "alkylcycloalkyl" means a cycloalkyl group linked to an alkyl group. The stated carbon number range includes the total number of carbons in the radical, unless otherwise specfically stated. This a C4-10 alkylcycloalkyl may contain from 1-7 carbon atoms in the alkyl group and from 3-9 carbon atoms in the ring, e.g., cyclopropylmethyl or cyclohexylethyL
The term "aryl" as used herein means an aromatic moiety containing the indicated number of carbon atoms, such as, but not limited to phenyl, indanyl or naphthyl. For example, C6-Io aryl refers to an aromatic moiety having from six to ten carbon atoms which may be in the form of a monocyclic or bicyclic structure. The term "haloaryl" as used herein refers to an aryl mono, di or tri substituted with one or more halogen atoms. The terms "alkylaryl", "arylalkyl" and "aralalkyl" mean an aryl group substituted with one or more alkyl groups. Unless the carbon range of each group is specified, the stated range applies to the entire substituent. Thus, a C7.H alkylaryl group many have from 1-8 carbon atoms in the alkyl group for a monocyclic aromatic and from 1-4 carbon atoms in the alkyl group for a fused aromatic. The attachment of the group to bonding site on the molecule can be either at the aryl group or the alkyl group. Unless a specific aryl radical is specified, e.g., fluoro-phenyl, or the radical is stated to be unsubstituted, the aryl radicals include those substituted with typical substituents known to those skilled in the art, e.g., halogen, hydroxy, carboxy, carbonyl, nitro, sulfo, amino, cyano, dialkylamino haloalkyl, CF3, haloalkoxy, thioalkyl, alkanoyl, SH, alkylamino, alkylamide, dialkylamide, carboxyester, alkylsulfone, alkylsulfonamide and alkyl(alkoxy)amine. Examples of alkylaryl groups include benzyl, butylphenyl and 1-naphthylmethyl. The term "alkanoyl" as used herein means straight or branched 1-oxoalkyl radicals containing the indicated number of carbon atoms and includes, for example, formyl, acetyl, 1-oxopropyl (propionyl), 2-methyl-l-oxopropyl, 1-oxohexyl and the like.
The term "alkylamide" as used herein means an amide mono-substituted with an alkyl, such as
Figure imgf000016_0001
The term "heterocycle", also referred to as "Het", as used herein means 7-12 membered bicyclic heterocycles and 5-7 membered monocyclic heterocycles. Preferred bicyclic heterocycles are 7-12 membered fused bicyclic ring systems (both rings share an adjacent pair of atoms) containing from one to four heteroatoms selected from nitrogen, oxygen and sulfur, wherein both rings of the heterocycle are folly unsaturated. The nitrogen and sulfur heteroatoms atoms may be optionally oxidized. The bicyclic heterocycle may contain the heteroatoms in one or both rings. Unless a specific heterocycle is specified, e.g., a fluorinated 7-12 membered bicyclic heterocycle, or the heterocycle is stated to be unsubstituted, the heterocycles include those substituted with typical substituents known to those skilled in the art. For example, the bicyclic heterocycle may also contain substituents on any of the ring carbon atoms, e.g., one to three substituents. Examples of suitable substituents include Cj-6 alkyl, C3-7 cycloalkyl, Cue alkoxy, C3-7 cycloalkoxy, halo-Ci- 6 alkyl, CF3, mono-or di- halo-Ci-6 alkoxy, cyano, halo, thioalkyl, hydroxy, alkanoyl, NO2, SH, , amino, Ci-6 alkylamino, di (C ι-β) alkylamino, di (C\-e) alkylamide, carboxyl, (Ci-β) carboxyester, Ci-β alkylsulfone, Ci-β alkylsulfonamide, Ci_6 alkylsulfoxide, di (Ci-β) alkyl(alkoxy)amine, Cβ-ioaryl, C7-I4 alky laryl, and a 5-7 membered monocyclic heterocycle. When two substituents are attached to vicinal carbon atoms of the bicyclic heterocycle, they can join to form a ring, e.g., a five, six or seven membered ring system containing up to two heteroatoms selecting from oxygen and nitrogen. Heterocycles may be attached to the parent molecular moiety through any substitutable carbon atom or nitrogen atom in the ring. Examples of bicyclic heterocycles include, but are not limited to, the following ring systems:
Figure imgf000016_0002
Preferred bicyclic heterocycles are those other than
Figure imgf000017_0001
wherein X and Y, together with the carbon atoms to which they are attached, form a ring selected from aryl and heteroaryl; X1 is selectd from CH and N, and Y1 and Y2 are independently selected from CH, O, S, and NH; wherein either ring system can be substituted or unsubstituted at any substitutable carbon or nitrogen atom in the ring.
Preferred monocyclic heterocycles are 5-7 membered saturated, partially saturated or fully unsaturated ring system (this latter subset also herein referred to as unsaturated heteroaromatic) containing in the ring from one to four heteroatoms selected from nitrogen; oxygen and sulfur, wherein the sulfur and nitrogen heteroatoms may be optionally oxidized. Unless a specific heterocycle is specified, e.g., a C1-6 alkoxy substituted 5-7 membered monocyclic heterocycle, or the heterocycle is stated to be unsubstituted, the heterocycles include those substituted with typical substituents known to those skilled in the art. For example, the monocyclic heterocycle may also contain substituents on any of the ring atoms, e.g., one to three substituents. Examples of suitable substituents include Ci-6 alkyl, C3-7 cycloalkyl, Ci_6 alkoxy, C3-7 cycloalkoxy, halo-Ci-g alkyl, CF3, mono-or di- halo-Ci-6 alkoxy, cyano, halo, thioalkyl, hydroxy, alkanoyl, NO2, SH, amino, Ci-β alkylamino, di (Ci-β) alkylamino, di (Cj-g) alkylamide, carboxyl, (Ci-e) carboxyester, Cj-6 alkylsulfone, C 1 -6 alkylsulfoxide, C 1.6 alkylsulfonamide, di (C 1 -β) alkyl(alkoxy)amine, Ce-io arylj C7_i4 alkylaryl and an additional 5-7 membered monocyclic heterocycle. The monocyclic heterocycle may be attached to the molecule, e.g. Ri in formula (I), at any atom in the ring.
Examples of monocyclic heterocycles include, but are not limited to, the following (and their tautomers):
Figure imgf000018_0001
Thos skilled in the art will recognize that the heterocycles used in the compounds of the present disclosure should be stable. Generally, stable compounds are those which can be synthesized, isolated and formulated using techniques known the those skilled in the art without degradation of the compound.
The term "substituent" with reference to an amino acid or amino acid derivative means a radical derived from the corresponding α-amino acid. For instance, the substituents methyl, iso-propyl, and phenyl represent the amino acids alanine, valine, and phenyl glycine, respectively. Where used in naming compounds of the present disclosure, the designations
"Pl', Pl, P2, P3 and P4", as used herein, map the relative positions of the amino acid residues of a protease inhibitor binding relative to the binding of the natural peptide cleavage substrate. Cleavage occurs in the natural substrate between Pl and Pl ' where the nonprime positions designate amino acids starting from the C-terminus end of the peptide natural cleavage site extending towards the N-terminus; whereas, the prime positions emanate from the N-terminus end of the cleavage site designation and extend towards the C-terminus. For example, Pl ' refers to the first position away from the right hand end of the C-terminus of the cleavage site (ie. N-terminus first position); whereas Pl starts the numbering from the left hand side of the C-terminus cleavage site, P2: second position from the C-terminus, etc.)(see Berger A. & Schechter L, Transactions of the Royal Society London series (1970), B257, 249-264].
The compounds of the present disclosure have the structure of formula (I):
Figure imgf000019_0001
wherein:
(a) R4 is H; C 1-6 alkyl, or C3-7 cycloalkyl each optionally substituted with halo; alkoxy; -C(O)-R5; C(O)-NHR5; or C(O)-OR5; wherein R5 is Ci-9 alkyl, optionally substituted with Ci.6 alkoxy, €3.7 cycloalkoxy, halo-Ci-6 alkoxy, cyano, halo, hydroxy, amino, Ci.6 alkylamino, di (Cr6) alkylamino, di (Ci-ό) alkylamide, carboxyl, (Cr6) carboxyester; C6-Io aryl; C7-I4 alkylaryl; heterocylclyl; OrC3-7 cycloalkyl;
(b) R6 is H, Cl-6 alkyl, or C3-7 cycloalkyl; (c) R3 and R'3 are each independently H or methyl;
(d) Q is a C3-9 saturated or unsaturated chain, optionally containing one to three heteroatoms independently selected from O or S(O)m; wherein m is 0, 1 or 2;
(e) W is OH, -O-Rl , or -NH-SO2-R2: wherein Rl is Cl -6 alkyl, unsaturated C3- 7 cycloalkyl, C6-14 aryl or C7-16 alkylaryl; and R2 is Cl-8 alkyl, C4-10 alkylcycloalkyl, unsubstituted C3-7 cycloalkyl; or R2 is cyclopropyl or cyclobutyl optionally substituted with C 1-4 alkyl or C7-16 alkylaryl, alkoxy, halo, haloalkyl, cyano, alkylcyano, or haloalkoxy;
(f) X is O, S5 SO, SO2, OCH2, CH2O or NH;
(g) R' is Het, C6-10 aryl or C7-14 alkylaryl, each optionally substituted with Ra; or C3-9 cycloalkyl or C 1-7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; and
(h) Ra is Cl-6 alkyl, C3-7 cycloalkyl, Cl-6 alkoxy, C3-7 cycloalkoxy, halo-Cl-6 alkyl, CF3, mono-or di- halo-Cl-6 alkoxy, cyano, halo, thioalkyl, hydroxy, alkanoyl, NO2, SH, , amino, Cl-6 alkylamino, di (Cl-6) alkylamino, di (Cl-6) alkylamide, carboxyl, (Cl-6) carboxyester, Cl-6 alkylsulfone, Cl-6 alkylsulfonamide, di (Cl-6) alkyl(alkoxy)amine, C6-10 aryl, C7-14 alkylaryl, or a 5-7 membered monocyclic heterocycle.
In one aspect, X is O5 S, OCH2, CH2O or NH. In another aspect, X is O or OCH2. In another aspect, X is O. In one aspect, R' is Het or C6-1O aryl, each optionally substituted with Ra. In another aspect, R' is Het. Often, the Het contains 1 or 2 nitrogen atoms and optionally a sulfur atom or an oxygen atom in the ring. In one aspect, the heterocycle is substituted with at least one of Ci -6 alkyl, Ci .6 alkoxy, halo, Ce aryl, and a 5-7 membered monocyclic heterocycle. In one aspect, R' is a bicyclic heterocycle containing 1 nitrogen atom and substituted with methoxy and at least one of a Ce aryl and a 5-7 membered monocyclic heterocycle. In another aspect, R' is a monocyclic heterocycle containing 1 or 2 nitrogen atoms and substituted with methoxy and at least one of a Ce aryl and a 5-7 membered monocyclic heterocycle.
In one aspect of the disclosure, R' has a structure selected from:
Figure imgf000020_0001
each optionally substituted with Ra.
In another aspect of the disclosure, R' has a structure selected from:
Figure imgf000021_0001
Figure imgf000021_0002
each optionally substituted with Ra.
In an aspect of the disclosure R' is selected from the following heterocycles;
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000023_0002
In another aspect of the disclosure X-IT is selected from the following;
Figure imgf000024_0001
Figure imgf000024_0002
In one aspect of the disclosure, Ra is C1-6 alkyl, C3-7 cycloalkyl, C1-6 alkoxy, C3-7 cycloalkoxy, halo-C1_6 alkyl, CF3, halo-Cue alkoxy, cyano, halo, thioalkyl, hydroxy , amino, Ci .6 alkylamino, di (Ci-β) alkylamino, di (Ci -β) alkylamide, carboxyl, (C1-6) carboxyester, C1-6 alky lsulfone, Cl-6 alkylsulfonamide, di (Cl-6) alkyl(alkoxy)amine, C6-io aryl, C7-I4 alkylaryl, or a 5-7 membered monocyclic heterocycle. In one aspect, Ra is C1-6 alkyl, C3-7 cycloalkyl, C1-6 alkoxy, C3-7 cycloalkoxy, halo-C1-.6 alkyl, CF3, halo-Ci-6 alkoxy, cyano, halo, di (C1-O) alkylamino, di (C1-6) alkylamide, C6-Io aryl, C7.14 alkylaryl, or a 5-7 membered monocyclic heterocycle. Ln one aspect, Ra is CL6 alkyl, C^.η cycloalkyl, Ci^ alkoxy, Ca-7 cycloalkoxy, halo-Ci-.6 alkyl, CF3, halo-C[.6 alkoxy, cyano, halo, hydroxy, amino, Ci.6 alkylamino, di (Cr6) alkylamino, di (Q-β) alkylamide. In one aspect, Ra is Ci-6 alkyl, C3.7 cycloalkyl, C1-6 alkoxy, halo-Ci-6 alkyl, halo, amino, Cβaryl, or a 5- 7 membered monocyclic heterocycle. In one aspect, Ra is Ci-6 alkoxy or halo. In one aspect, Ra is methoxy or chloro.
In one aspect of the disclosure, -X-R' has a structure other than the following structure:
Figure imgf000025_0001
wherein:
(a) R7 is H, halo, Cl-6 alkyl, Cl-6 alkoxy, C3-6 cycloalkoxy, phenyl optionally substituted with Cl -3 alkyl, C 1-3 alkoxy or halo, or heterocycle optionally substituted with from one to three of halo, cyano, trifluoromethyl, nitro, Cl-6 alkyl, Cl-6 alkoxy, amido, Cl-6 alkanoylamino or amino; and
(b) R8 is H, halo, CF3, Cl-6 alkoxy, C3-6 cycloalkoxy or C 1-3 alkyl.
In one aspect, W is -NH-SO2-R2: wherein R2 is C1-8 alkyl, C4-10 alkylcycloalkyl, unsubstituted C3-7 cycloalkyl; or R2 is cyclopropyl or cyclobutyl optionally substituted with Ci-4 alkyl or C7-i6 alkylaryl. In one aspect, R2 is Ci-8 alkyl, C4-io alkylcycloalkyl or unsubstituted C3-7 cycloalkyl. In one aspect, R2 is cyclopropyl or cyclobutyl.
In one aspect, Q is a Cs-7 saturated or unsaturated chain optionally containing one to three heteroatoms independently selected from O or S(O)n,; wherein m is 0, 1 or 2. In one aspect, Q is a Ce saturated or unsaturated chain optionally containing one to three heteroatoms independently selected from O or S(O)m; wherein m is 0, 1 or 2. In one aspect, Q is unsaturated. In one aspect, Q has six carbon atoms. In one aspect, Q has a structure selected from:
Figure imgf000025_0002
Figure imgf000026_0001
wherein P is a C3 saturated chain optionally containing one heteroatom independently selected from O, S(0)m; wherein m is 0, 1 or 2.
In one aspect, R2 is cyclopropyl substituted in the Cl position with C 1-4 alkyl or C7-16 alkylaryl, or the C2 or C3 position with Cl-4 alkyl. In one aspect, R2 is cyclobutyl substituted in the Ci position with C1-4 alkyl or C7- 1 β alkylaryl, or the C2, C3 or C4 position with CM alkyl. In one aspect, R4 is -C(O)-R5, C(O)-NHR5 or C(O)- OR5; wherein R5 is Ci-6 alkyl optionally substituted with halo, alkoxy, or cyano. In one aspect, R5 is Cj.6 alkyl optionally substituted with halo. In one aspect, R5 is Ci-6 alkyl.
In one aspect, R3 and R' 3 are each independently H or methyl. In one aspect, R6 is H or Ci-6 alkyl.
In one aspect of the disclosure, the compounds of the present disclosure have the structure of formula (II):
Figure imgf000027_0001
wherein:
(a) R4 is -C(O)-R5, C(O)-NHR5 or C(O)-OR5; wherein R5 is C]-9 alkyl optionally substituted with Ci-6 alkoxy, cyano, halo;
(b) Q is a C5-7 saturated or unsaturated chain optionally containing one to three heteroatoms independently selected from O or S(O)m; wherein m is 0, 1 or 2;
(c) W is NH-SO2-R2, wherein R2 is Ci-8 alkyl, C4-io alkylcycloalkyl, unsubstituted C3.7 cycloalkyl, or cyclopropyl or cyclobutyl optionally substituted with Ci-4 alkyl or C7-16 alkylaryl;
(d) X is O, S, OCH2, CH2O or NH;
(e) R' is Het, Cβ-io aryl or C7-H alkylaryl, each optionally substituted with Ra; or C3.9 cycloalkyl or C1.7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; and (f) Ra is Ci-6 alkyl, C3.7 cycloalkyl, C1.6 alkoxy, C3-7 cycloalkoxy, halo-Ci-.g alkyl, CF3, halo-Ci.6 alkoxy, cyano, halo, thioalkyl, hydroxy , amino, Ci-O alkylamino, di (Ci-β) alkylamino, di (Ci-β) alkylamide, carboxyl, (Q-β) carboxyester, Cj.6 alkylsulfone, Ci-β alkylsulfonamide, di (Ci-e) alkyl(alkoxy)amine, C6-Io aryl, C7-H alkylaryl, or a 5-7 membered monocyclic heterocycle. In an aspect of the disclosure relating to the compounds of formula (II), (a) R4 is -C(O)-R5, C(O)-NHR5 or C(O)-OR5,- wherein R5 is Ci-9 alkyl optionally substituted halo; (b) Q has a structure selected from;
Figure imgf000028_0001
Figure imgf000029_0001
wherein P is a C3 saturated chain optionally containing one heteroatom independently selected from O, S(O)n,; wherein m is 0, 1 or 2;
(a) W is NH-SO2-R2, wherein R2 is C1-8 alkyl, C3-? cycloalkyl,
(b) X is O, or OCH2;
(c) R' is Het, Cό-io ary.1 or C7-I4 alkylaryl, each optionally substituted with Ra; and Ra is C1-6 alkyl, C3-7 cycloalkyl, C^alkoxy, C3-7 cycloalkoxy, halo-Ci-.β alkyl, halo- Ci-6alkoxy, cyano, halo, di (Q-e) alkylamino.
In another aspect of the disclosure relating to the compounds of formula (II),
(a) R4 is -C(O)-R5, C(O)-NHR5 or C(O)-OR5; wherein R5 is Ci-9 alkyl;
(b) Q has a structure selected from;
Figure imgf000029_0002
Figure imgf000030_0001
(c) W is NH-SO2-R2, wherein R2 is cyclopropyl;
(d) X is O;
(e) R' is Het or Cβ-io aryl each optionally substituted with Ra; and Ra is methoxy or chloro.
In one aspect of the disclosure, the compounds of the present disclosure have the structure of formula (III):
Figure imgf000030_0002
wherein: (a) R4 is -C(O)-R5, C(O)-NHR5 or C(O)-OR5; wherein R5 is C1-6 alkyl;
(b) Q has a structure selected from;
Figure imgf000031_0001
(c) W is NH-SO2-R2, wherein R2 is cyclopropyl; and (d) R' is Het or Ce- 10 aryl.
In an aspect of the disclosure relating to the compounds of formula (III), (a) Q has a structure selected from:
Figure imgf000032_0001
(b) R' has a structure selected from the following
Figure imgf000032_0002
Figure imgf000033_0001
each R' is optionally substituted with Ra; wherein Ra is selected from alkoxy, c.yano, cycloalkoxy, halo, or dialkyamino.
In an aspect of the disclosure relating to the compounds of formula (III), R1 has a structure selected from the following
Figure imgf000033_0002
Figure imgf000034_0001
each R' is optionally substituted with Ra; wherein Ra is selected from alkoxy or halo. The compounds of the present disclosure, which contain a basic moiety, can form salts by the addition of a pharmaceutically acceptable acid. The acid addition salts are formed from a compound of formula (I) and a pharmaceutically acceptable inorganic acid, including but not limited to hydrochloric, hydrobromic, hydroiodic, sulfuric, phosphoric, or organic acid such as/?-toluenesulfonic, methanesulfonic, acetic, benzoic, citric, malonic, fumaric, maleic, oxalic, succinic, sulfamic, or tartaric. Thus, examples of such pharmaceutically acceptable salts include chloride, bromide, iodide, sulfate, phosphate, methanesulfonate, citrate, acetate, malonate, fumarate, sulfamate, and tartrate.
Salts of an amine group may also comprise quaternary ammonium salts in which the amino nitrogen carries a suitable organic group such as an alkyl, alkenyl, alkynyl or aralkyl moiety. Compounds of the present disclosure, which are substituted with an acidic group, may exist as salts formed through base addition. Such base addition salts include those derived from inorganic bases which include, for example, alkali metal salts (e.g. sodium and potassium), alkaline earth metal salts (e.g. calcium and magnesium), aluminum salts and ammonium salts. In addition, suitable base addition salts include salts of physiologically acceptable organic bases such as trimethylamine, triethylamine, morpholine, pyridine, piperidine, picoline, dicyclohexylamine, N,N' -dibenzylethylenediamine, 2-hydroxyethylamine, bis-(2-hydroxyethyl)amine, tri-(2-hydroxyethyl)amine, procaine, dibenzylpiperidine, N-benzyl-β-phenethylamine, dehydroabietylamine, N5N '-bishydroabietylamine, glucamine, N-methylglucamine, collidine, quinine, quinoline, ethylenediamine, ornithine, choline, N,N'-benzylphenethylamine, chloroprocaine, diethanolamine, diethylamine, piperazine, tris(hydroxymethyl)aminomethane and tetramethylammonium hydroxide and basic amino acids such as lysine, arginine and N-tnethylglutamine. These salts may be prepared by methods known to those skilled in the art.
Certain compounds of the present disclosure, and their salts, may also exist in the form of solvates with water, for example hydrates, or with organic solvents such as methanol, ethanol or acetonitrile to form, respectively, a methanolate, ethanolate or acetonitrilate. The present disclosure includes each solvate and mixtures thereof. In addition, compounds of the present disclosure, or a salt or solvate thereof, may exhibit polymorphism. The present disclosure also encompasses any such polymorphic form. Asymmetric centers exist in the compounds of the present disclosure. For example, the compounds may include Pl cyclopropyl element of formula
Figure imgf000035_0001
wherein Ci and C2 each represent an asymmetric carbon atom at positions 1 and 2 of the cyclopropyl ring. Not withstanding other possible asymmetric centers at other segments of the compounds, the presence of these two asymmetric centers means that the compounds can exist as racemic mixtures of diastereomers, such as the diastereomers wherein R2 is configured either syn to the amide or syn to the carbonyl as shown below.
Figure imgf000036_0001
Figure imgf000036_0002
It should be understood that the disclosure encompasses all stereochemical isomeric forms, or mixtures thereof, which possess the ability to inhibit HCV protease. The enantiomers may be resolved by methods known to those skilled in the art, for example, by formation of diastereoisomeric salts which may be separated by crystallization, gas-liquid or liquid chromatography, selective reaction of one enantiomer with an enantiomer-specific.reagent. It will be appreciated that where the desired enantiomer is converted into another chemical entity by a separation technique, then an additional step is required to form the desired enantiomeric form. Alternatively, specific enantiomers may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents, or by converting one enantiomer into the other by asymmetric transformation.
Certain compounds of the present disclosure may also exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers. The present disclosure includes each conformational isomer of these compounds and mixtures thereof. Certain compounds of the present disclosure may exist in zwitterionic form and the present disclosure includes each zwitterionic form of these compounds and mixtures thereof.
The starting materials useful to synthesize the compounds of the present disclosure are known to those skilled in the art and can be readily manufactured or are commercially available .
The compounds of the present disclosure can be manufactured by methods known to those skilled in the art. The following methods set forth below are provided for illustrative purposes and are not intended to limit the scope of the claimed disclosure. For example, compounds of the present disclosure having the structure of formula (I) can be synthesized, as shown in scheme 1. It will be recognized that it may be preferred or necessary to prepare such a compound in which a functional group is protected using a conventional protecting group then to remove the protecting group to provide a compound of the present disclosure. The details concerning the use of protecting groups in accordance with the present disclosure are known to those skilled in the art.
As shown in scheme 1, intermediates of the present disclosure such as dipeptide 1, can be used for the preparation of compounds of formula (I). In the first step of this process the Boc protected nitrogen of 1 is deprotected using an acid such as HCl in a solvent such as ether, to provide the corresponding free amine 2. Amine 2 can be subsequently coupled to amino acid 3 using a coupling agent such as HATU in a solvent such as dichloromethane to provide the tripeptide intermediate 4. It should be noted that in some cases intermediates like 3 are commercially available, and alternatively such compounds can be readily prepared in racemic or chiral fashion by methods known in the art. A key transformation in the construction of compounds of formula (I) is the macrocyclization process wherein intermediates of general structure 4 are converted into intermediates of general structure 5. In the general example cited, the conversion of intermediate 4 into 5 can be affected by an intramolecular olefin metathesis reaction. This class of reactions is well established in the art and as such, a number of olefln-metathesis-catalysts have been developed and are commercially available. For example the conversion of diene 4 to macrocycle 5 could be affected by the treatment of 4 with a sufficient quantity of Grubb's first-generation olefin metathesis catalyst, in a solvent such as dichloromethane or dichloroethane. In some examples for the conversion of 4 to 5, it may be necessary to heat the reaction mixture in order to effect this cyclization process. Intermediate 5 is then coverted to compounds of formula (I) such as 7 by a two step process. In the first step of this process, the ester functionality of intermediate 5 is hydrolyzed to the corresponding carboxylic 6. This transformation can be accomplished by a saponification reaction wherein 5 is treated with a base such as lithium hydroxide in a mixture of THF5 methanol and water. The resulting acid 6 can be converted to a compound of formula (I) by a simple coupling reaction with a sulfonamide derivative as shown. For example, it is well established in the art that treatment of a carboxylic acid like 6, with CDI in a solvent such as methylene chloride, generates in situ a reactive intermediate which when treated with a sulfonamide provides for 7, a compound of formula (I).
Scheme 1
Figure imgf000039_0001
Figure imgf000039_0002
Figure imgf000039_0003
An additional process wherein compounds of formula (I) can be prepared is outlined below (Scheme 2). Therein, the P2* functionality, defined herein as the functional group attached to the proline C4-moiety (XR"), is incorporated after the P1-P3 macrocyclization step. However, it should be noted that the process of incorporating of P2* into the molecule can be executed at any suitable stage of the synthesis. In the present, nonlimiting scheme, (Scheme 2) the proline substituent X is protected using a suitable protecting group. This group is then carried through several steps in the synthesis as shown, and removed after the cyclization process to provide an intermediate like 5, which is then converted into a compound of formula (I) (6)-
Scheme 2
Figure imgf000040_0001
In the construction of compounds of formula (I), the PT terminus is incorporated into the molecules using one of the general processes outlined above and described in more detail below. In some examples the P 1 " elements, that is the cycloalkylsulfonamides or alkyl sulfonamides, are commercially available or can be prepared from the corresponding alkyl- or cycloalkyl-sulfonyl chloride by treating said sulfonyl chloride with ammonia . Alternatively, these sulfonamides can be synthesized using the general process outlined in Scheme 3. Therein commercially available 3-chloropropylsulfonyl chloride (1) is converted to a suitable protected sulfonamide as for example by treatment with tert-butyl amine. The sulfonamide 2 obtained is then converted to the corresponding cycloalkylsulfonamide 3 by treatment with two equivalents of a base such as butyl lithium in a solvent such as THF at low temperature. The resulting cycloalkylsulfonamide can be deprotected by treatment with an acid to provide the desired unprotected cycloalkylsulfonamide 4. Said PV fragment 4 can be incorporated into compounds of formula (I). Additionally, the cycloalkyl ring of intermediates like 4 can be further functionalized. For example, treatment of intermediate 3 with a base such as butyl lithium followed by the addition of an electrophile such as an alkyl halide should provide intermediates like 5, wherein the Cl position of the cycloalkyl ring is functionalized. Reactions of this type can be conducted in solvents such as THF. In such a reaction it may be necessary to add two or more equivalents of base to intermediate 3. Moreover, the temperature of such a reaction will likely need to be carefully monitored wherein the THF solution of 3 is cooled to -78C prior to the addition of base and this is described in detail herein. Scheme 3
Figure imgf000041_0001
As an alternative to the t-butyl protecting group used in the above scheme (eg. intermediate 2 of scheme 3) a Boc group can be employed as shown below (Scheme 4). Said Boc group can be incorporated by treatment of an intermediate like 2 with Boc anhydride in the presence of a base such as triethylamine in conjunction with catalytic DMAP. The acylsulfonamide 3 obtained is then converted to the corresponding cycloalkylacylsulfonamide 4 by treatment with two equivalents of a base such as butyl lithium in a solvent such as THF at low temperature. The resulting cycloalkylacylsulfonamide 4 can be deprotected by treatment with an acid to provide the desired unprotected cycloalkylsulfoamide. Said PT fragment can be incorporated into compounds of formula (I).
Scheme 4
Figure imgf000042_0001
In the preparation of compounds of formula (I)3 dipeptide intermediates like 2 shown below can be prepared by the coupling of hydroxyproline derivative 1 with cyclopropyl amino acid B as shown. This coupling reaction can be carried out using reagents such as HATU or HBTU and in solvents such as methylene chloride or DMF or THF.
Figure imgf000042_0002
Intermediate B can be synthesized as shown in scheme 5.
Scheme 5
Figure imgf000043_0001
Treatment of commercially available, or readily synthesized imine 1 with 1,4- dihalobutene 2 in presence of a base provides the imine 3. Acid hydrolysis of 3 then provides B as a mixture of diastereoisomers. It is preferred that for compounds 3 and B that the vinyl group is syn to the ester. The amine moiety of B can protected using a Boc group to provide the fully protected amino acid 4a/4b. This intermediate is a racemate, a 1: 1 mixture of enantiomers, and each enantiomer is shown in the above scheme. Racemate 4a/4b can be resolved by an enzymatic process wherein the ester moiety of 4 is cleaved to provide the corresponding carboxylic acid. Without being bound to any particular theory, it is believed that this reaction is selective in that one of the enantiomers, that is 4b, with the absolute stereochemistry designated (IS, 2R) undergoes the reaction at a much greater rate than its mirror image, 4a, providing for a kinetic resolution of racemate 4a/4b. Hence, in the course of this enzyme catalyzed ester cleavage, 4b will be readily converted to the corresponding acid 5, whereas 4a will remain as unreacted starting material. Once the reaction is terminated, the carboxylic acid 5 and recovered starting material 4a, can be separated by rountine methods such as aqueous extraction methods or chromatography. As shown below, intermediate 4a can be readily converted into compounds of formula (I) by the methods described herein. For example, removal of the Boc group from intermediate 4a can be accomplished by subjecting 4a to an acid such as HCl in a solvent such as ether, to provide the corresponding amine hydrochloride 6. Intermediate 6 can then be coupled to a functionalized proline moiety 1 to provide the P1-P2 dipeptide 2. Intermediates like 2 can be converted to compounds of formula (I) by the methods described herein.
Figure imgf000044_0001
Compounds of formula (I) can also be converted into other compounds of formula (I) as described herein. An example of such a process is shown in Scheme 6, wherein a compound of formula (I) (1) which bears a Boc group at the P4 position is converted into a compound of formula (I) (3) wherein said compound bears a urea group at the P4 position. The conversion of 1 to 3 can be carried out in a two step process the first of which is the conversion of 1 to amine 2 by treatment of 1 with an acid such as TFA in a solvent such as methylene chloride. The resulting amine TFA salt can be treated with an isocyanate in the presence of one equivalent of base to provide a compound of formula (I) (3) wherein the P3 moiety is capped with a urea. As previously noted one skilled in the art will recognize that intermediate 2 can be used as starting material for the preparation of compounds of formula (I) wherein the P3 group is capped with an amide or a carbamate. The construction of said compounds of formula (I) can be achieved using standard conditions for the formation of said P4 functionalities from amines. Scheme 6
Figure imgf000045_0001
Figure imgf000045_0002
One skilled in the art would recognize that the incorporation of P2* into the molecule can be carried out at any stage in the assembly of the peptide backbone. This is illustrated below (Scheme 7) for the conversion of intermediates like 1, 3 or 5 into compounds of formula (I).
Scheme 7
Figure imgf000046_0001
Figure imgf000046_0002
Figure imgf000046_0003
The present disclosure also provides compositions comprising a compound of the present disclosure, or a pharmaceutically acceptable enantiomer, diastereomer, salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier. Pharmaceutical compositions of the present disclosure comprise a therapeutically effective amount of a compound of the disclosure, or a pharmaceutically acceptable enantiomer, diastereomer, salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier, with a pharmaceutically acceptable carrier, e.g., excipient, or vehicle diluent.
The active ingredient, i.e., compound, in such compositions typically comprises from 0.1 weight percent to 99.9 percent by weight of the composition, and often comprises from about 5 to 95 weight percent.
Thus, in one aspect of the disclosure, there is provided a composition comprising the compound of formula (I) and a pharmaceutically acceptable carrier. Preferably, the composition further comprises a compound having anti-HCV activity. As used herein, the term "anti-HCV activity" means the compound is effective to inhibit the function of a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, IMPDH and a nucleoside analog for the treatment of an HCV infection. Often, the other compound having anti-HCV activity is effective to inhibit the function of target in the HCV life cycle other than the HCV NS3 protease protein.
In one preferred aspect, the compound having anti-HCV activity is an interferon. Preferably, the interferon is selected from the group consisting of interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, lymphoblastiod interferon tau.
In another aspect of the disclosure, the compound having anti-HCV activity is selected from the group consisting of interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
In one preferred aspect of the disclosure, the composition comprises a compound of the disclosure, an interferon and ribavirin. In another preferred aspect of the disclosure, the compound having anti-HCV activity is a small molecule compound. As used herein, the term "small molecule compound" means a compound having a molecular weight of less than 1,500 daltons, preferably less than 1000 daltons. Preferably, the small molecule compound is effective to inhibit the function of a target selected from the group consisting of HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, inosine monophophate dehydrogenase ("IMPDH") and a nucleoside analog for the treatment of an HCV infection.
Certain illustrative HCV inhibitor compounds which can be administered with the compounds of the present disclosure include those disclosed in the following publications: WO 02/04425 A2 published January 17, 2002, WO 03/007945 Al published January 30, 2003, WO 03/010141 A2 published February 6, 2003, WO 03/010142 A2 published February 6, 2003, WO 03/010143 Al published February 6, 2003, WO 03/000254 Al published January 3, 2003, WO 01/32153 A2 published May 10, 2001, WO 00/06529 published February 10, 2000, WO 00/18231 published April 6, 2000, WO 00/10573 published March 2, 2000, WO 00/13708 published March 16, 2000, WO 01/85172 Al published November 15, 2001, WO 03/037893 Al published May 8, 2003, WO 03/037894 Al published May 8, 2003, WO 03/037895 Al published May 8, 2003, WO 02/100851 A2 published December 19, 2002, WO 02/100846 Al published December 19, 2002, EP 1256628 A2 published November 13, 2002, WO 99/01582 published January 14, 1999, WO 00/09543 published February 24, 2000.
Table 1 below lists some illustrative examples of compounds that can be administered with the compounds of this disclosure. The compounds of the disclosure can be administered with other anti-HCV activity compounds in combination therapy, either jointly or separately, or by combining the compounds into a composition.
TABLE 1
Figure imgf000048_0001
Figure imgf000049_0001
The pharmaceutical compositions of this disclosure may be administered orally, parenterally or via an implanted reservoir. Oral administration or administration by injection are preferred. In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, and intralesional injection or infusion techniques.
When orally administered, the pharmaceutical compositions of this disclosure may be administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added. Other suitable carriers for the above noted compositions can be found in standard pharmaceutical texts, e.g. in "Remington's Pharmaceutical Sciences", 19th ed., Mack Publishing Company, Easton, Perm., 1995.
The pharmaceutical compositions can be prepared by known procedures using well-known and readily available ingredients. The compositions of this disclosure may be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures well known in the art. In making the compositions of the present disclosure, the active ingredient will usually be admixed with a carrier, or diluted by a carrier, or enclosed within a carrier which may be in the form of a capsule, sachet, paper or other container. When the carrier serves as a diluent, it may be a solid, semi-solid or liquid material which acts as a vehicle, excipient or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, beadlets, lozenges, sachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols, (as a solid or in a liquid medium), soft and hard gelatin capsules, suppositories, sterile injectable solutions, sterile packaged powders and the like. Further details concerning the design and preparation of suitable delivery forms of the pharmaceutical compositions of the disclosure are known to those skilled in the art. Dosage levels of between about 0.01 and about 1000 milligram per kilogram ("mg/kg") body weight per day, preferably between about 0.5 and about 250 mg/kg body weight per day of the compounds of the 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. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. As the skilled artisan will appreciate, lower or higher doses than those recited above may be required. Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the infection, the patient's disposition to the infection and the judgment of the treating physician. Generally, treatment is initiated with small dosages substantially less than the optimum dose of the peptide. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. In general, 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.
When the compositions of this disclosure comprise a combination of a compound of the 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 100%, and more preferably between about 10 and 80% of the dosage normally administered in a monotherapy regimen.
When these compounds or their pharmaceutically acceptable enantiomers, diastereomers, salts, solvates or prodrugs are formulated together with a pharmaceutically acceptable carrier, the resulting composition may be administered in vivo to mammals, such as man, to inhibit HCV NS 3 protease or to treat or prevent HCV virus infection.
Accordingly, another aspect of this disclosure provides methods of inhibiting HCV NS3 protease activity in patients by administering a compound of the present disclosure or a pharmaceutically acceptable enantiomer, diastereomer, salt or solvate thereof. hi one aspect of the disclosure, there is provided a method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of the compound of the disclosure, or a pharmaceutically acceptable enantiomer, diastereomer, solvate, prodrug or salt thereof.
Preferably, the method of administering the compound is effective to inhibit the function of the HCV NS3 protease protein, hi a preferred aspect, the method further comprises administering another compound having anti-HCV activity (as described above) prior to, after or concurrently with a compound of the disclosure. The compounds of the disclosure may also be used as laboratory reagents. Compounds may be instrumental in providing research tools for designing of viral replication assays, validation of animal assay systems and structural biology studies to further enhance knowledge of the HCV disease mechanisms. Further, the compounds of the present disclosure are useful in establishing or determining the binding site of other antiviral compounds, for example, by competitive inhibition. The compounds of this disclosure may also be used to treat or prevent viral contamination of materials and therefore reduce the risk of viral infection of laboratory or medical personnel or patients who come in contact with such materials, e.g., blood, tissue, surgical instruments and garments, laboratory instruments and garments, and blood collection or transfusion apparatuses and materials.
Further, the compounds and compositions of the disclosure can be used for the manufacture of a medicament for treating HCV infection in a patient.
EXAMPLES
The specific examples that follow illustrate the syntheses of the compounds of the present disclosure, and are not to be construed as limiting the scope of the claims which follow. The methods may be adapted to variations in order to produce compounds embraced by this disclosure but not specifically disclosed. Further, variations of the methods to produce the same compounds in somewhat different manner will also be evident to one skilled in the art.
Chemical abbreviations commonly used to identify chemical compounds disclosed herein include Bn: benzyl; Boc: tert-butyloxycarbonyl (Me3COC(O)); BSA: bovine serum albumin; CDI: carbonyldiimidazole; DBU: 1,8-diazabicy- clo[5.4.0]-undec-7-ene; CH2Cl2=DCM: methylene chloride; TBME: tert-butyl methyl ether; DEAD: diethylazodicarboxylate; DIAD: diisopropylazodicarboxylate; DIEA: diisopropylethylamine; DIPEA: diisopropylethylamine; 4-DMAP: 4- dimethylaminopyridine; DCC: 1,3-dicyclohexylcarbodiimide; DMF: dimethylfoπnamide; DMSO: dimethylsulfoxide; DPPA: diphenylphosphoryl azide; Et: ethyl; EtOH: ethanol; EtOAc: ethyl acetate; Et2O: diethyl ether; Grubb's Catalyst: bis(tricyclohexylphosphine)benzylidene ruthenium (IV) dichloride; Grubb's 2nd Generation Catalyst: tricyclohexylphosphine[l,3- bis(2,4,6-trimethylphenyl)-4,5- dihydroimidazol-2-ylidene][benzylidene]ruthenium (IV) dichloride; HATU: [O-(7- azabenzotriazol-1 -yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate; HBTU: [O-(l H-benzotriazol- 1 -yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate; HOBT, 1 -hydroxybenzotriazole; HOAT, l-hydroxy-7-azabenzotriazole; HPLC: high performance liquid chromatography; MS: mass spectrometry; Me: methyl; MeOH: methanol; NMM: N-methylmorpholine; NMP: N-methylpyrrolidine; Pr: propyl; PPA: polyphosphoric acid; TBAF: tetra-n-butylammonium fluoride; 1,2-DCE or DCE: 1,2-dichloroethane; TFA: trifluoroacetic acid; THF: tetrahydrofuran.
Solution percentages express a weight to volume relationship, and solution ratios express a volume to volume relationship, unless stated otherwise. Nuclear magnetic resonance (NMR) spectra were recorded either on a Bruker 300, 400 or 500 megahertz (MHz) spectrometer; the chemical shifts (δ) are reported in parts per million. Flash chromatography was carried out on silica gel (SiO2) according to Still's flash chromatography technique (J. Org. Chem. 1978, 43, 2923). Liquid Chromatography (LC) data were recorded on a Shimadzu LC-IOAS liquid chromatograph using a SPD-I OAV UV- Vis detector and Mass Spectrometry (MS) data were determined with a Micromass Platform for LC in electrospray mode (ES+). Solution percentages express a weight to volume relationship, and solution ratios express a volume to volume relationship, unless stated otherwise. Nuclear magnetic resonance (NMR) spectra were recorded either on a Bruker 300, 400 or 500 MHz spectrometer; the chemical shifts (δ) are reported in parts per million.
The examples, compounds and chemical intermediates of the present disclosure, described in the following examples, were prepared according to the following methods. Example 1:
Preparation ofracemic (lR,2S)/(lS,2R)-l~amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride (Method A and Method B)
Figure imgf000054_0001
The named compound was made racemic by each of the following methods A and B.
Figure imgf000054_0002
Glycine ethyl ester hydrochloride (303.8 g, 2.16 mole) was suspended in tert- butylmethyl ether (1.6 L). Benzaldehyde (231 g, 2.16 mole) and anhydrous sodium sulfate (154.6 g, 1.09 mole) were added and the mixture cooled to 0 °C using an ice- water bath. Triethylamine (455 mL, 3.26 mole) was added dropwise over 30 min and the mixture stirred for 48 h at rt. The reaction was then quenched by addition of ice- cold water (1 L) and the organic layer was separated. The aqueous phase was extracted with tert-butylmethyl ether (0.5 L) and the combined organic phases washed with a mixture of saturated aqueous NaHCOs (1 L) and brine (1 L). The solution was dried over MgSO4, concentrated in vacuo to afford 392.4 g of the N- benzyl imine product as a thick yellow oil that was used directly in the next step. 1H NMR (CDCl3, 300 MHz) 6 1.32 (t, J=I. ,1 Hz, 3H), 4.24 (q, J=I.1 Hz, 2H), 4.41 (d, J=Ll Hz, 2H), 7.39-7.47 (m, 3H), 7.78-7.81 (m, 2H), 8.31 (s, IH). Preparation of racemic N-Boc-(lR,2S)/(lS,2R)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester
Figure imgf000054_0003
To a suspension of lithium /ert-butoxide (84.06 g, 1.05 mol) in dry toluene (1.2 L), was added drop wise a mixture of the N-benzyl imine of glycine ethyl ester (100.4 g, 0.526 mol) and _>τ3rts-l,4-dibromo-2-butene (107.0 g, 0.500 mol) in dry toluene (0.6 L) over 60 min. After completion of the addition, the deep red mixture was quenched by addition of water (1 L) and /er/-butylmethyl ether (TBME, 1 L). The aqueous phase was separated and extracted a second time with TBME (1 L). The organic phases were combined, 1 N HCl (1 L) was added and the mixture stirred at room temperature for 2 h. The organic phase was separated and extracted with water (0.8 L). The aqueous phases were then combined, saturated with salt (700 g), TBME (1 L) was added and the mixture cooled to 0 0C. The stirred mixture was then basified to pH 14 by the dropwise addition of 10 N NaOH, the organic layer separated, and the aqueous phase extracted with TBME (2 x 500 mL). The combined organic extracts were dried (MgSO4) and concentrated to a volume of IL. To this solution of free amine, was added BOC2O or di-før/-butyldicarbonate (131.0 g, 0.6 mol) and the mixture stirred 4 days at rt. Additional di-fer£-butyldicarbonate (50 g, 0.23 mol) was added to the reaction, the mixture refluxed for 3 h, and was then allowed cool to room temperature overnight. The reaction mixture was dried over MgSO4 and concentrated in vacuo to afford 80 g of crude material. This residue was purified by flash chromatography (2.5 Kg of SiO2, eluted with 1% to 2% MeOH/CH2Cl2) to afford 57 g (53%) of racemic
Figure imgf000055_0001
vinylcyclopropane carboxylic acid ethyl ester as a yellow oil which solidified while sitting in the refrigerator: 1H NMR (CDCl3, 300 MHz) δ 1.26 (t, J=7.1 Hz, 3H), 1.46 (s, 9H), 1.43-1.49 (m, IH), 1.76-1.82 (br m, IH), 2.14 (q, J=8.6 Hz, IH), 4.18 (q, J=7.2 Hz, 2H), 5.12 (ddJ=10.3, 1.7 Hz, IH), 5.25 (br s, IH), 5.29 (dd, J=I 7.6, 1.7 Hz, IH), 5.77 (ddd, J=17.6, 10.3, 8.9 Hz, IH); MS m/z 254.16 (M-I).
Preparation of Racemic (1R,2S)/(1S,2R) l-amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride
Figure imgf000055_0002
7V-Boc-(li?,2iS)/(15',2i?)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester (9.39 g, 36.8 mmol) was dissolved in 4 N HCl/dioxane (90 ml, 360 mmol) and was stirred for 2 h at rt. The reaction mixture was concentrated to supply (li?,25)/(15,2i?)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride in quanitative yield (7 g, 100%). 1H NMR (methanol-d4) δ 1.32 (t, J=7.l, 3H), 1.72 (dd, J=10.2, 6.6 Hz, IH), 1.81 (dd, J=8.3, 6.6 Hz, IH), 2.38 (q, J=8.3 Hz, IH), 4.26-4.34 (m, 2H), 5.24 (dd, 10.3, 1.3 Hz, IH) 5.40 (d, J=17.2, IH), 5.69-5.81 (m, IH).
Method B
Preparation ofRacemic N-Boc-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride
Figure imgf000056_0001
To a solution of potassium tert-butoxide (11.55 g, 102.9 mmol) in THF (450 mL) at -78 0C was added the commercially available iV,iV-dibenzyl imine of glycine ethyl ester (25.0 g, 93.53 mmol) in THF (112 mL). The reaction mixture was warmed to 0 0C, stirred for 40 min, and was then cooled back to -78 0C. To this solution was added trows1- l,4-dibromo-2-butene (20.0 g, 93.50 mmol), the mixture stirred for 1 h at 00C and was cooled back to — 78°C. Potassium tert-butoxide (11.55 g, 102.9 mmol) was added, the mixture immediately warmed to 0°C, and was stirred one more hour before concentrating in vacuo. The crude product was taken up in Et2O (530 mL), IN aq. HCl solution (106 mL, 106 mmol) added and the resulting biphasic mixture stirred for 3.5 h at rt. The layers were separated and the aqueous layer was washed with Et2θ (2x) and basified with a saturated aq. NaHCO3 solution. The desired amine was extracted with Et2O (3x) and the combined organic extract was washed with brine, dried (MgSO4), and concentrated in vacuo to obtain the free amine. This material was treated with a 4N HCl solution in dioxane (100 mL, 400 mmol) and concentrated to afford (li?,2/S)/(15',2/?)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride as a brown semisolid (5.3 g, 34% yield) identical to the material obtained from procedure A, except for the presence of a small unidentified aromatic impurity (8%). Example 2
Resolution ofN-Boc-(lR,2S)/(lS,2R)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester
Figure imgf000057_0001
Resolution A
To an aqueous solution of sodium phosphate buffer (0.1 M, 4.25 liter ("L"), pH 8) housed in a 12 Liter jacked reactor, maintained at 390C, and stirred at 300 rpm was added 511 grams of Alcalase 2.4L (about 425 mL) (Novozymes North America Inc.). When the temperature of the mixture reached 39°C, the pH was adjusted to 8.0 by the addition of a 50% NaOH in water. A solution of the racemic iV-Boc-(li?,2_S)/(lS',2/0- l-amino-2-vinylcyclopropane carboxylic acid ethyl ester (85g) in 850 mL of DMSO was then added over a period of 40 min. The reaction temperature was then maintained at 4O0C for 24.5h during which time the pH of the mixture was adjusted to 8.0 at the 1.5h and 19.5h time points using 50% NaOH in water. After 24.5h, the enantio-excess of the ester was determined to be 97.2%, and the reaction was cooled to room temperature (26°C) and stirred overnight (16h) after which the enantio- excess of the ester was determined to be 100%. The pH of the reaction mixture was then adjusted to 8.5 with 50% NaOH and the resulting mixture was extracted with MTBE (2 x 2 L). The combined MTBE extract was then washed with 5% NaHCO3 (3 x 100 mL), water (3 x 100 mL), and evaporated in vacuo to give the enantiomerically pure N-Boc-(li2,2S)/-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester as light yellow solid (42.55 g; purity: 97% @ 210 nm, containing no acid; 100% enantiomeric excess ("ee"). The aqueous layer from the extraction process was then acidified to pH 2 with 50% H2SO4 and extracted with MTBE (2 x 2 L). The MTBE extract was washed with water (3 x 100 mL) and evaporated to give the acid as light yellow solid (42.74 g; purity: 99% @ 210 nm, containing no ester).
Figure imgf000058_0001
Figure imgf000058_0002
Resolution B
To 0.5 mL 100 mM Heps»Na buffer (pH 8.5) in a well of a 24 well plate (capacity: 10 ml/well), 0.1 mL of Savinase 16.0L (protease from Bacillus clausii) (Novozymes North America Inc.) and a solution of the racemic N-Boc- ( 1 R,2S)/( 1 S,2R)-l -amino-2-vinylcyclopropane carboxylic acid ethyl ester (10 mg) in 0.1 mL of DMSO were added. The plate was sealed and incubated at 250 rpm at 400C. After 18h, enantio-excess of the ester was determined to be 44.3% as following: 0.1 mL of the reaction mixture was removed and mixed well with 1 mL ethanol; after centrifugation, 10 microliter ("μl") of the supernatant was analyzed with the chiral HPLC. To the remaining reaction mixture, 0.1 mL of DMSO was added, and the plate was incubated for additional 3 days at 250 rpm at 400C, after which four mL of ethanol was added to the well. After centrifugation, 10 μl of the supernatant was analyzed with the chiral HPLC and enantio-excess of the ester was determined to be 100%. Resolution C
To 0.5 ml 100 mM Heps»Na buffer (pH 8.5) in a well of a 24 well plate (capacity: 10 mL/well), 0.1 ml of Esperase 8.0L, (protease from Bacillus halodurans) (Novozymes North America Inc.) and a solution of the racemic 7V-Boc- (1R,2S)/(IS,2R)-1 -amino-2-vinylcyclopropane carboxylic acid ethyl ester (10 mg) in 0.1 mL of DMSO were added. The plate was sealed and incubated at 250 rpm at 400C. After 18 hour, enantio-excess of the ester was determined to be 39.6% as following: 0.1 mL of the reaction mixture was removed and mixed well with 1 mL ethanol; after cenrifugation, 10 μl of the supernatant was analyzed with the chiral HPLC. To the remaining reaction mixture, 0.1 mL of DMSO was added, and the plate was incubated for additional 3 days at 250 rpm at 400C, after which four mL of ethanol was added to the well. After centrifugation, 10 μl of the supernatant was analyzed with the chiral HPLC and enantio-excess of the ester was determined to be 100%. Sample analysis was carried out in the following manner: 1) Sample preparation: About 0.5 ml of the reaction mixture was mixed well with 10 volume of EtOH. After centrifugation, 10 μl of the supernatant was injected onto HPLC column. 2) Conversion determination:
Column: YMC ODS A3 4.6 x 50 mm, S-5 μm Solvent: A, 1 mM HCl in water; B, MeCN
Gradient: 30% B for 1 min; 30% to 45% B over 0.5 min; 45% B for 1.5 min; 45% to 30% B over 0.5 min. Flow rate: 2 ml/min UV Detection: 210 nm Retention time: acid, 1.2 min; ester, 2.8 min.
3) Enantio-excess determination for the ester: Column: CHIRACEL OD-RH, 4.6 x 150 mm, S-5 μm Mobile phase: MeCN/50 mM HClO4 in water (67/33) Flow rate: 0.75 ml/min. UV Detection: 210 nm. Retention time: (15',2i?)-l-amino-2-vinylcyclopropane carboxylic acid 5.2 min;
Racemate (li?,25)/(15',2i?)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester 18.5 min and 20.0 min;
(112,25)- l-amino-2-vinylcyclopropane carboxylic acid ethyl ester 18.5 min. Resolution D 5 L of 0.3 M sodium phosphate buffer (pH 8) was maintained at 38°C in a 20,
Liter jacked reactor, stirred at 130 rpm. Four liters of Alcalase 2.4L (Novozymes North America Inc.) and 1 liter of DI water were added to the reactor. When temperature of the mixture closed to 380C, pH was adjusted to 7.8 with 10 N NaOH. A solution of the racemic iV-Boc-(li?,2»S)/(15',2i?)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester (500 grams) in 5 liters DMSO was added to the reactor over a period of 1 hour via an addition funel. The reaction temperature was then adjusted to 48°C. After 21 hours, enantio-excess of the ester reached 99.3%. Heating was stopped at 24 hour and the reaction was slowly cooled down to room temperature (about 250C) and stirred overnight. pH of the reaction mixture was adjusted to 8.5 with 10 N NaOH and the mixture was extracted with MTBE (2 x 4 L). The combined MTBE extract was washed with 5% NaHCO3 (3 x 400 ml) and water (3 x 400 ml), and evaporated to give enantiomerically pure iV-Boc-(l/?,25)/-l-amino- 2-vinylcyclopropane carboxylic acid ethyl ester as light yellow crystal (259 g; purity: 96.9% @ 210 nm, containing no acid; 100%ee). Resolution E
10 L of 0.1 M sodium phosphate buffer (pH 8) was maintained at 4O0C in a 20 Liter jacked reactor, stirred at 360 rpm. 1.5 liters of Alcalase 2.4L (Novozymes North America Inc.) was added to the reactor. When temperature of the mixture closed to 380C, pH was adjusted to 8.0 with IO N NaOH. A solution of the racemic iV-Boc-(li?,2Sy(lS,2ϋ)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester (200 grams) in 2 liters DMSO was added to the reactor over a period of 1 hour via an addition funel. The reaction temperature was then adjusted to 4O0C. After 3 hours, pH was adjusted to 8.0 with IO N NaOH. After 21 hours, the reaction was cooled down to 250C. pH of the reaction mixture was adjusted to 8.5 with IO N NaOH and the mixture was extracted with MTBE (2 x 5 L). The combined MTBE extract was washed with 5% NaHCO3 (3 x 500 ml) and water (3 x 200 ml), and evaporated to give 110 gram of yellow oil. The oil was set at room temperature under house vacuum and gave enantiomerically pure iV-Boc-(l/?,25)/-l-amino-2- vinylcyclopropane carboxylic acid ethyl ester as colorless long rod crystal (101 g; purity: 97.9% @ 210 nm, containing no acid; 100%ee).
The crystal structure enantiomerically pure iV-Boc-(l/?,2,S)/-l-amino-2- vinylcyclopropane carboxylic acid ethyl ester has been characterized by single crystal analysis (X-ray NB#: 52795-093, refcode: 634592N1). The absolute configuration is not established for lack of a known chiral center or heavier atom(s). A chain structure along the crystallographic α-axis is formed via intermolecular hydrogen bonding between the amide group and the carbonyl oxygen atom (N...O 3.159 A). Structure of iV-Boc-(li?,2iS)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester:
Figure imgf000061_0001
Crystal Data: Experimental:
Chemical formula: C13H21N1O4 Crystallization
Crystal system: Orthorhombic Crystal source: MTBE
Space Group: P2,2i2! Crystal description: Colorless rod a - 5.2902(I) A a = 90° Crystal size (mm): 0.12 X 0.26 X 0.30 b = 13.8946(2) A /9 = 90° Data Collection c = 19.9768(3) A γ = 90° Temperature (K): 293
V- 1468.40(4) A3 0max (°): 65.2 (Cu Ka)
Z = A dx = 1.155 g em"3 No. of reflections measured: 7518
No. of reflections for lattice parameters: 6817 No. of independent reflections: 2390 (Rin,=
0.0776) θ range for lattice parameters (°): 2.2-65.2 No. of observed reflections (/> 2 σ): 2284
Absorption coefficient (mm"1): 0.700 Absorption correction {Tmin-Tmax): 0.688-1.000
Resolution F
5 L of 0.2 M sodium borate buffer (pH 9) was maintained at 45°C in a 20 liter jacked reactor, stirred at 400 rpm. Three liter of DI water and four liters of Savinase 16L, type EX (Novozymes North America Inc.) were added to the reactor. When temperature of the mixture closed to 450C, pH was adjusted to 8.5 with IO N NaOH. A solution of the racemic iV-Boc-(li?,2iS)/(15',2i2)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester (200 grams) in 2 liters DMSO was added to the reactor over a period of 40 min, via an addition funel. The reaction temperature was then adjusted to 480C. After 2 hours, pH was adjusted to pH 9.0 with IO N NaOH. At 18 hour, enantio-excess of the ester reached 72%, pH was adjusted to 9.0 with IO N NaOH. At 24 hour, temperature was lowered to 35°C. At 42 hour, temperature was raised to 480C and pH was adjusted to 9.0 with IO N NaOH. Heating was stopped at 48 hour and the reaction was slowly cooled down to room temperature (about 250C) and stirred overnight. At 66 hour, pH of the reaction mixture was 8.6. The mixture was extracted with MTBE (2 x 4 L). The combined MTBE extract was washed with 5% NaHCO3 (6 x 300 ml) and water (3 x 300 ml), and evaporated to give give enantiomerically pure iV-Boc-( 1.^,25)/- l-amino-2-vinylcyclopropane carboxylic acid ethyl ester as light yellow crystal (101A g; purity: 95.9% @ 210 nm, containing no acid; 98.6%ee). Example 3
Step 1: Preparation of ethyl l(R)-amino-2(S)-vinylcyclopropane carboxylate hydrochloride
Figure imgf000063_0001
Ethyl l(R)-tert-butoxycarbonylamino-2(S)-vinylcyclopropanecarboxylate (8.5 g, 33.3 mmol) was stirred under an N2 atmosphere with 200 mL of 4N HCl/dioxane (Aldrich) at rt for 3h. The solvent was removed under reduced pressure keeping the temperature below 40 C. This gave 6.57 g (-100%) of ethyl l(R)-amino-2(S)- vinylcyclopropanecarboxylate hydrochloride as a light tan solid. 1H NMR (300 MHz, CD3OD) δ 1.31 (t, J=7.0 Hz9 3 H), 1.69-1.82 (m, 2 H), 2.38 (q, J=8.8 Hz, 1 H), 4.29 (q, J=7.0 Hz5 2 H)5 5.22 (d5 J=10.3 Hz5 1 H)5 5.40 (d5 J=17.2 Hz5 1 H)5 5.69-5.81 (m, l H). MS rø/z l56 (M++l).
Step 2: Preparation of ethyl 1 (R)-[I -tert-butoxycarbonyl-4 (R)-hydroxypyrrolidine- 2(S)-carboxamido]-2(S)-vinylcyclopropanecarboxylate
Figure imgf000063_0002
A stirred slurry of Boc-L-4-hydroxyproline (iV-Boc (25'54i?)-hydroxyproline)
(10 g, 43.3 mmol) in 400 mL of methylene chloride was treated sequentially with N- methyl morpholine (9.3 mL, 84.7 mmol), HATU (19.5 g, 51.3 mmol), and ethyl l(R)-amino-2(S)-vinylcyclopropanecarboxylate hydrochloride (9.1 g, 47.5 mmol). The gold homogeneous solution was stirred at rt under N2 for 18h, and then concentrated in vacuo to give a brown oil. This was partitioned between ethyl acetate and sat. aq. NaHCCh. The organic phase was washed with brine, dried (MgSO4), and concentrated in vacuo to give 15 g (94%) of ethyl 1 (R)-[I -tert-butoxycarbonyl-4(R)- hydroxypyrrolidine-2(S)-carboxamido]-2(S)-vinylcyclopropanecarboxylate as a off- white solid: LC-MS (Xterra HPLC column: 3.0x50 mm length. Gradient: 100% Solvent A/0% Solvent B to 0% Solvent A/100% Solvent B. Gradient time: 3 min. Hold time: 1 min. Flow rate: 5 mL/min. Detector Wavelength: 220 nm. Solvent A: 10% MeOH / 90% H2O / 0.1% TFA. Solvent B: 10% H2O / 90% MeOH / 0.1% TFA.) (Retention time: 2.09 min), MS m/z 369 (M++l).
Step 3: Preparation of ethyl l(R)-[4(R)-hydroxypyrrolidine-2(S)-carboxamido]-2(S)- vinylcyclopropanecarboxylate hydrochloride.
Figure imgf000064_0001
A stirred slurry of ethyl 1(R)-[I -tert-butoxycarbonyl-4(R)- hydroxypyrrolidine-2(S)-carboxamido] -2(S)-vinylcyclopropanecarboxylate (5.0 g, 13.6 mmol) was treated with 4N HCl/dioxane (20 mL) for 3 h. The reaction mixture was concentrated in vacuo to give 4.5 g (97%) of ethyl 1(R)-[4(R)- hydroxypyrrolidine-2(S)-carboxamido]-2(S)-vinylcyclopropanecarboxylate hydrochloride as a white solid: 1H NMR (300 MHz, CD3OD) δ 1.26 (t, 7=7.14 Hz, 3 H), 1.46 (dd, /=9.70, 5.31 Hz, 1 H)5 1.80 (dd, J=8.23, 5.31 Hz, 1 H)3 2.00 - 2.15 (m, 1 H), 2.18 - 2.30 (m, 1 H), 2.45 (dd, J=13.36, 7.50 Hz, 1 H), 3.36 - 3.48 (m, 1 H), 4.11 - 4.24 (m, 2 H)5 4.44 (dd, J=I 0.25, 7.68 Hz, 1 H), 4.58 - 4.65 (m, 1 H), 4.84 - 4.94 (m, 1 H), 5.17 (d, J=1.83 Hz, 1 H), 5.27 - 5.42 (m, 1 H), 5.67 - 5.89 (m, 1 H). Example 4
Preparation of Cyclopropylsulfonamide Methods A and B Method A:
Figure imgf000064_0002
To a solution of 100 mL of THF cooled to 00C was bubbled in gaseous ammonia until saturation was reached. To this solution was added a solution of 5 g (28.45 mmol) of cyclopropylsulfonyl chloride (purchased from Array Biopharma) in 50 mL of THF, the solution warmed to rt overnite and stirred one additional day. The mixture was concentrated until 1-2 mL of solvent remained, applied on to 30 g plug OfSiO2 (eluted with 30% to 60% EtOAc/Hexanes) to afford 3.45g (100%) of cyclopropyl sulfonamide as a white solid. 1H NMR (Methanol-α^) δ 0.94-1.07 (m, 4H), 2.52-2.60 (m, IH); 13C NMR (methanol^) δ 5.92, 33.01.
Method B:
Step 1: Preparation ofN-tert-Butyl-(3-chloro)propylsulfonamide
Figure imgf000065_0001
tert-Butylamine (3.0 mol, 315.3 tnL) was dissolved in THF (2.5 L). The solution was cooled to — 200C. 3-Chloropropanesulfonyl chloride (1.5 mol, 182.4 mL) was added slowly. The reaction mixture was allowed to warm to rt and stirred for 24 h. The mixture was filtered, and the filtrate was concentrated in vacuo. The residue was dissolved in CH2CI2 (2.0 L). The resulting solution was washed with 1 N HCl (1.0 L), water (1.0 L), brine (1.0 L) and dried over Na2SO4. It was filtered and concentrated in vacuo to give a slightly yellow solid, which was crystallized from hexane to afford the product as a white solid (316.0 g, 99%).
1H NMR (CDCl3) δ 1.38 (s, 9H), 2.30-2.27 (m, 2H), 3.22 (t, J= 7.35 Hz, 2H), 3.68 (t, J = 6.2 Hz, 2H), 4.35 (b, IH). Step 2: Preparation of Cyclopropanesulfonic acid tert-butylamide
Figure imgf000065_0002
To a solution of N-tert-butyl-(3-chloro)propylsulfonamide (2.14 g, 10.0 mmol) in THF (100 mL) was added n-BuLi (2.5 M in hexane, 8.0 mL, 20.0 mmol) at -780C. The reation mixture was allowed to warm up to room temperature over period of 1 h. The volatiles were removed in vacuo. The residue was partitioned between EtOAC and water (200 mL, 200 mL). The separated organic phase was washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was recrystallized from hexane to yield the desired product as a white solid (1.O g5 56%). 1H NMR (CDCl3) δ 0.98-1.00 (m, 2H), 1.18-1.19 (m, 2H), 1.39 (s, 9H), 2.48-2.51 (m, IH), 4.19 (b, IH). Step 3: Preparation of cyclopropylsulfonamide
Figure imgf000065_0003
A solution of cyclopropanesulfonic acid tert-butylamide (110.0 g, 0.62 mol) in TFA (500 mL) was stirred at room temperature for 16 h. The volatile was removed in vacuo. The residue was recrystallized from EtOAC/hexane (60 mL/240 mL) to yield the desired product as a white solid (68.5 g, 91%). 1H NMR (DMSO-d6) δ 0.84-0.88 (m, 2H), 0.95-0.98 (m, 2H), 2.41-2.58 (m, IH), 6.56 (b, 2H). Example 5
Preparation ofN-tert-butyl-(l-methyl)cyclopropylsϊilfonamide.
Figure imgf000066_0001
Step Ia Preparation ofN-tert-butyl-(3-chloro)propylsulfonamide.
Figure imgf000066_0002
As shown above.
Step Ib. Preparation ofN-tert-Butyl-(l-methyl)cyclopropylsulfonamide.
Figure imgf000066_0003
A solution of iV-tørt-butyl-(3-chloro)propylsulfonamide (4.3 g, 20 mmol) was dissolved in dry THF (100 mL) and cooled to — 78 0C. To this solution was added n- BuLi (17.6 mL, 44 mmol, 2.5 M in hexane) slowly. The dry ice bath was removed and the reaction mixture was allowed to warm to rt over a period of 1.5h. This mixture was then cooled to - 78°C, and a solution of n-BuLi (20 mmol, 8 mL, 2.5 M in hexane) was added. The reaction mixture was warmed to rt, recooled to -78 0C over a period of 2 h and a neat solution of methyl iodide (5.68 g, 40 mmol) added. The reaction mixture was allowed to warm to rt overnight, quenched with saturated NH4Cl (10O mL) at rt. It was extracted with EtOAc (100 mL). The organic phase was washed with brine (100 mL), dried (MgSO4), and concentrated in vacuo to give a yellow oil which was crystallized from hexane to afford the product as a slightly yellow solid (3.1 g, 81%): 1H NMR (CDCl3) δ 0.79 (m, 2H), 1.36 (s, 9H), 1.52 (m, 2H), 1.62 (s, 3H), 4.10 (bs, IH). Step Ic: Preparation ofl-methylcyclopropylsulfonamide
Figure imgf000067_0001
A solution of iV-^er/-butyl-(l-methyi)cyclopropylsulfonamide (1.91 g, 10 mrαol) was dissolved in TFA (30 rnL), and the reaction mixture stirred at rt for 16 h. The solvent was removed in vacuo to give a yellow oil which was crystallized from EtOAc / hexane (1 : 4, 40 mL) to yield Example 3, 1-methylcyclopropylsulfonamide, as a white solid (1.25 g, 96%): 1H NMR (CDCl3) δ 0.84 (m, 2H), 1.41 (m, 2H), 1.58 (s, 3H), 4.65 (bs, 2H). Anal. Calcd. For C4H9NO2S: C5 35.54; H, 6.71; N, 10.36. Found: C, 35.67; H, 6.80; N5 10.40. Example 6
Preparation of I -Ben∑ylcyclopropylsulfonamide
Figure imgf000067_0002
Steps Ib: Preparation ofN-tert-Butyl-ζl-benzytycyclopropyl-sulfonamide.
Figure imgf000067_0003
This compound was obtained in 60% yield using the procedure described for the synthesis of 7V-tert-butyl-(l-methyl)cyclopropylsulfonamide except 1.05 equivalents of benzyl bromide were used, followed by trituration with 10% EtOAc in hexane: 1H NMR (CDCl3) δ 0.92 (m, 2H)5 1.36 (m, 2H), 1.43 (s, 9H), 3.25 (s, 2H)5 4.62 (bs, IH), 7.29-7.36 (m5 5H). Steps Ic: Preparation ofl-Benzylcyclo-propylsulfonamide
Figure imgf000067_0004
This compound l-benzylcyclopropylsulfonamide, was obtained in 66% yield fromN-ter/-butyl(l~benzyl)cyclopropylsulfonamide using the procedure described for the synthesis of 1-methylcyclopropylsulfonamide, followed by recrystallization from the minimum amount of 10% EtOAc in hexane: 1H NMR (CDCl3) δ 0.90 (m, 2H)5 1.42 (m, 2 H)5 3.25 (s, 2 H), 4.05 (s5 2 H), 7.29 (m, 3 H), 7.34 (m, 2 H); 13C NMR (CDCl3) δ 11.1, 36.8, 41.9, 127.4, 128.8, 129.9, 136.5. Example 7
Preparation of 1-Propylcyclopropylsulfonamide
Figure imgf000068_0001
Steps Ib: Preparation of N-tert-Butyl-(l -benzyl) cyclopropyl-sulfonamide.
Figure imgf000068_0002
This compound was prepared using the process desribed for the preparation of 1-methylcyclopropylsulfonamide except propyl halide was utilized in place of methyl iodide in the second step of this process. Example 8 Preparation ofN-tert-Butyl-(l-allyl)cyclopropylsulfonamide.
Figure imgf000068_0003
This compound, N-ter?-Butyl-(l-allyl)cyclopropylsulfonamide, was obtained in 97% yield according to the procedure described in the synthesis ofN-tert-Butyl-(l- methyl)cyclopropylsul-fonamide except 1.25 equivalents of allyl bromide were used as electrophile. The compound was taken directly into the next reaction without purification: 1H NMR (CDCl3) δ 0.83 (m, 2H), 1.34 (s, 9H)5 1.37 (m, 2H)3 2.64 (d, J = 7.3 Hz, 2H), 4.25 (bs, IH)5 5.07-5.10 (m, 2H), 6.70-6.85 (m, IH). Preparation of ' 1-allylcyclopropylsulfonamide.
Figure imgf000068_0004
This compound, 1-allylcyclopropylsulfonamide, was obtained in 40% yield fromΛT-ter/-butyl-(l-allyl)cyclopropylsulfonamide according to the procedure described in the synthesis of 1-Methylcyclopropylsulfonamide. The compound was purified by column chromotography over SiO2 using 2% MeOH in CH2Cl2 as the eluent: 1H NMR (CDCl3) δ 0.88 (m, 2 H), 1.37 (m, 2 H), 2.66 (d, J=7.0 Hz, 2 H), 4.80 (s, 2 H), 5.16 (m, 2 H), 5.82 (m, 1 H); 13C NMR (CDCl3) δ 11.2, 35.6, 40.7, 119.0, 133.6. Example 9
Preparation ofN-tert-Butyl-[l - (1 -hydroxy)cyclohexyl]-cyclopropylsulfonamide.
Figure imgf000069_0003
This compound was obtained in 84% yield using to the procedure described for the synthesis of iV-/erf-Butyl-(l-methyl)cyclopropylsul-fonamide except 1.30 equivalents of cyclohexanone were used, followed by recrystallization from the minimum amount of 20% EtOAc in hexane: 1H NMR (CDCl3) δ 1.05 (m, 4H), 1.26 (m, 2H), 1.37 (s, 9H), 1.57-1.59 (m, 6H), 1.97 (m, 2H), 2.87 (bs, IH), 4.55 (bs, IH). Example 10 Preparation ofl-(l-cyclohexenyl)cyclopropyl-sulfonamide.
Figure imgf000069_0001
This compound, l-(l-cyclohexenyl)-cyclopropylsulfonamide was obtained in 85% yield fromN-tert-butyl-[l-(l-hydroxy)cyclohexyl]-cyclopropylsulfonamide 1H NMR (DMSO-de) δ 0.82 (m, 2 H), 1.28 (m, 2 H), 1.51 (m, 2 H), 1.55 (m, 2 H), 2.01 (ss 2 H), 2.16 (S5 2 H), 5.89 (s, 1 H), 6.46 (s, 2 H); 13C NMR (DMSOd6) δ 11.6, 21.5, 22.3, 25.0, 27.2, 46.9, 131.6, 132.2; LR-MS (ESI): 200 (M+-I). Example 11 Preparation ofN-tert-Butyl-(l -benzoyl)cyclopropyl-sulfonamide.
Figure imgf000069_0002
This compound was obtained in 66% yield using the procedure described for the synthesis of iV-ter*-Butyl-(l -methyl) cyclopropylsulfonamide except 1.2 equivalents of methyl benzoate was used as the electrophile. The compound was purified by column chromatography over SiO2 using 30% to 100% CH2Cl2 in hexane: 1H NMR (CDCl3) δ 1.31 (s, 9H), 1.52 (m, 2H), 1.81 (m, 2H), 4.16 (bs, IH), 7.46 (m9 2H), 7.57 (m, IH), 8.05 (d, J= 8.5 Hz, 2H). Preparation of l-benzoylcyclo-propylsulfonamide.
Figure imgf000070_0001
This compoundl-benzoylcyclopropyl-sulfonamide, was obtained in 87% yield from N-?ert-butyl(l-benzoyl)cyclopropylsul-fonamide using the procedure described for the synthesis of 1-Methylcyclopropylsulfonamide, followed by recrystallization from the minimum amount of EtOAc in hexane: 1H NMR (DMSO- d6) δ 1.39 (m, 2 H), 1.61 (m, 2 H)5 7.22 (s, 2 H), 7.53 (t, J=7.6 Hz5 2 H)5 7.65 (t, J=7.6 Hz, 1 H)5 8.06 (d, J=8.2 Hz, 2 H); 13C NMR (DMSO-d6) δ 12.3, 48.4, 128.1, 130.0, 133.4, 135.3, 192.0. Example 12
Preparation ofN-tert-Butyl~(l-phenylaminocarboxy)-cyclopropylsulfonamide
Figure imgf000070_0002
This compound was obtained in 42% yield using the procedure described for the synthesis of iV-ϊert-Butyl-(l-methyl)cyclopropylsulfonarnide using 1 equivalent of phenylisocyanate, followed by recrystallization from the minimum amount of
EtOAc in hexane 1H NMR (CDCl3) δ 1.38 (s, 9H)5 1.67-1.71 (m, 4H), 4.30 (bs, IH), 7.10 (t, J= 7.5 Hz, IH), 7.34 (t, J= 7.5 Hz, 2H), 7.53 (t, J= 7.5 Hz, 2H). Example 13 .
Preparation of cyclopropylsulfonylamine tert-butyl carbamate, a key intermediate in the preparation of Cl -substituted cyclopropylsulfonamides
Figure imgf000070_0003
Step 1: Preparation of 3-chloropropylsulfonamide
Figure imgf000070_0004
A solution of 3-chloropropanesulfonyl chloride (55 g, 310.7 mmol) was dissolved in THF (200 mL) and added dropwise over 30 minutes to a solution of NH4OH (200 mL) cooled to 00C. The reaction mixture was warmed to room temperature, stirred 1 hour, and the aqueous layer partioned multiple time with dichloromethane (4 x 500 mL). The combined dichloromethane layer was washed with IN HCl (150 mL), water (150 mL), dried over MgSO4, filtered, and concentrated in vacuo. The crude solid was recrystallized from the minimum amount of dichloromethane in hexanes to afford 3-chloropropylsulfonamide as a white solid (45.3 g, 93%). 1H NMR (CDCl3) δ 2.34 (m, 2H), 3.32 (t, J=7.3 Hz, 2H), 3.70 (t, J=6.2 Hz, 2H), 4.83 (s, 2H); 13C NMR (CDCl3) δ 27.10, 42.63, 52.57. Step2: Preparation of 3-chloropropylsulfonylamine tert-butylcarbamate
Figure imgf000071_0001
To a solution of 3-chloropropylsulfonamide (30.2 g, 191.5 mmol), triethylamine (30.2 mL, 217.0 mmol), and 4-DMAP (2.4Og5 19.6 mmol) in dichloromethane (350 mL) cooled to 0 0C was added slowly dropwise a solution of di-ført-butyldicarbonate (47.2g, 216.9 mmol) in dichloromethane (250 mL) over 30 minutes. The reaction mixture was allowed to warm to room temperature, stirred an additional 3 hours and was pardoned with IN HCl (300 mL), water (300 mL), brine (300 mL), dried over MgSO4, filtered, and concentrated in vacuo to afford the crude product. This material was triturated with 70 mL of 5% dichloromethane in hexanes to afford 3-chloropropylsulfonylamine fert-butylcarbamate as an offwhite solid (47.2 g, 96%): 1H NMR (CDCl3) δ 1.51 (s, 9H), 2.33 (m, 2H), 3.60 (t, J=7.3 Hz, 2H), 3.68 (t, j=6.21 Hz, 2H); 13C NMR (CDCl3) δ 26.50, 27.95, 42.37, 50.40, 84.76, 149.53. Step 3: Preparation of cyclopropylsulfonylamine tert-butyl carbamate
Figure imgf000071_0002
A solution of n-butyl lithium (74.7 mL, 119.5 mmol, 1.6M in hexane) was dissolved in dry THF (105 mL) and cooled to -78 0C under a Argon atmosphere. To this solution was added a solution of 3-chloropropylsulfonylamine tert- butylcarbamate (14 g, 54.3 mmol) in dry THF (105 mL) dropwise over 20-30 minutes. The dry ice bath was removed and the reaction mixture was allowed to warm to room temperature over a period of 2 hours. The reaction mixture was quenched with glacial acetic acid (3.4 mL), concentrated in vacuo, and partitioned between dichloromethane (100 mL) and water (100 mL). The organic phase was washed with brine (100 mL), dried (MgSO4), filtered, and concentrated in vacuo to afford the cyclopropylsulfonylamine tert-huXy\ carbamate as a waxy off-white solid (12.08 g, 100%): 1H NMR (CDCl3) δ 1.10 (m, 2H), 1.34 (m, 2H)5 1.50 (s, 9H), 2.88 (m, IH), 7.43 (s, IH). 13C NMR (CDCl3) δ 6.21, 28.00, 31.13, 84.07, 149.82. Example 14 Preparation of l-methoxy-methylcyclopropy-sulfonamide
Figure imgf000072_0001
Step 1: Preparation ofl-methoxymethylcyclopropylsulfonylamine tert- butylcarbamate
Figure imgf000072_0002
To a solution of cyclopropylsulfonylamine ter/-butyl carbamate (1.0 g, 4.5 mmol) dissolved in THF (30 mL) cooled to -78 0C, was added n-butyl lithium (6.4 mL, 10.2 mmol, 1.6M in hexane) and the reaction mixture was stirred for 1 hour. To this solution was added a neat solution of chloromethyl methyl ether (0.40 mL, 5.24 mmol), and the mixture was slowly allowed to warm to room temperature overnight. The solution pH was adjusted to 3 using IN aqueous HCl and was then extracted with ethyl acetate (4 x 50 mL portions). The combined extracts were dried (MgSO4), filtered, and concentrated to afford 1-methoxymethylcyclopropylsulfonylamine tert- butylcarbamate, as a waxy solid (1.20 g, 100%) which was taken directly into the next reaction without further purification: 1H NMR (CDCl3) δ 1.03 (m, 2H), 1.52 (s, 9H), 1.66 (m, 2H), 3.38 (s, 3H), 3.68 (s, 2H), 7.54 (s, IH); 13C NMR (CDCl3) δ 11.37, 28.29, 40.38, 58.94, 73.43, 83.61, 149.57. Step 2: Preparation of 1-meihoxymethylcyclopropy sulfonamide
Figure imgf000072_0003
A solution of l-methoxymethylcyclopropylsulfonylamine fert-butylcarbamate
(1.14 g, 4.30 mmol) was dissolved in a solution of 50%TFA/dichloromethane (30 mL) and was stirred stirred at room temperature for 16 hours. The solvent was removed in vacuo and the residue chromatographed over 80g of S1O2 (eluting with 0% to 60% ethyl acetate/hexanes to l-methoxymethylcyclopropylsulfonamide as a white solid (0.55 g, 77% overall over two steps): 1H NMR (CDCl3) δ 0.95 (m, 2H), 1.44 (m, 2H), 3.36 (s, 3H), 3.65 (s, 2H), 4.85 (s, 2H); 13C NMR (CDCl3) δ 11.17, 40.87, 59.23, 74.80; LRMS m/z 183 (M+H-NH4). Example 15
Preparation of 1-cyclopropylmethylcyclopropylsulfonamide
Figure imgf000073_0001
Step 1: Preparation of 1-cyclopropylmethylcyclopropylsulfonylamine tert- - butylcarbamate
Figure imgf000073_0002
1 -Cyclopropylmethylcyclopropylsulfonylamine tert-butylcarbamate was obtained in 92% yield according to the procedure described in the synthesis of 1- methoxymethylcyclopropylsulfonylamine ført-butylcarbamate, except 1.10 equivalents of cyclopropylmethyl bromide were used as electrophile. The compound was taken directly into the next reaction without purification: 1H NMR (CDCl3) δ 0.10 (m, 2H), 0.51 (m, 2H), 0.67 (m, IH)5 1.10 (m, 2H), 1.49 (s, 9H), 1.62 (m, 2H), 1.87 (d, J=7.0 Hz, 2H). Step 2: Preparation ofl-cyclopropylmethylcyclopropylsulfonamide
Figure imgf000073_0003
This compound was obtained in 65% yield from 1- cyclopropylmethylcyclopropylsulfonylamine terf-butylcarbamate according to the procedure described for the synthesis of l-methoxymethylcyclopropylsulfonamide. The compound was purified by column chromotography over SiO2 using 0% to 60% ethyl acetate in hexanes as the eluent: 1H NMR (CDCl3) δ 0.15 (m, 2H), 0.51 (m, 2H), 1.01 (m, 2H), 1.34 (m, 3H), 1.86 (d, J=7.0 Hz, 2H), 4.83 (s, 2H); 13C NMR (CDCl3) δ 4.65, 7.74, 11.26, 35.62, 41.21; LRMS m/z 193 (M++NH4). Example 16
Preparation ofl-propylcarbamoylcyclopropanesulfonamide
Figure imgf000074_0001
Step 1: Preparation of l-propylcarbamoylcyclopropanesulfonamide tert- butylcarbamate
Figure imgf000074_0002
This compound was obtained in a crude 100% yield according to the procedure described for the synthesis of 1-methoxymethylcyclopropylsulfonylamine rerr-butyl-carbamate except that 1.10 equivalents of n-propyl isocyanate was used as the electrophile. The compound was taken directly into the next reaction without purification: 1H NMR (CDCl3) δ 0.10 (m, 2H), 0.51 (m, 2H), 0.67 (m, IH), 1.10 (m,
2H)5 1.49 (s, 9H), 1.62 (m, 2H)5 1.87 (d, J=7.0 Hz52H).
Step 2: Preparation ofl-propylcarbamoylcyclopropanesulfonamide
Figure imgf000074_0003
This compound was obtained in an optimized 50% yield from 1 - propylcarbamoylcyclopropanesulfonamide fer/-butylcarbamate according to the procedure described for the synthesis of 1-methoxymethylcyclopropylsulfonamide, except that no chromatography was used as the material was recrystallized from the minimum amount of dichloromethane/hexanes: 1H NMR (CDCl3) δ 0.15 (m, 2H)5 0.51 (m5 2H), 1.01 (m, 2H), 1.34 (m, 3H)5 1.86 (d, J=7.0 Hz5 2H)5 4.83 (s, 2H); 13C NMR (CDCl3) δ 4.65, 7.74, 11.26, 35.62, 41.21; LRMS m/z 193 (M++NH4). Example 17 Preparation ofl-fS^-dimethylisoxazol^ylJcarbamoylcyclopropanesulfonamide
Figure imgf000074_0004
Step 1: Preparation of l-(3,5-dimethylisoxazol-4- yl)carbamoylcyclopropanesulfonamide tert-butylcarbamate
Figure imgf000075_0001
This compound was obtained in a crude 100% yield according to the procedure described for the synthesis of l-methoxymethylcyclopropylsulfonylamine tørr-butylcarbamate except that 1.20 equivalents of 3,5-dimethylisoxazole-4- isocyanate was used as the electrophile. The compound was taken directly into the next reaction without purification.
Step 2: Preparation of l-(3,5-dimethylisoxazol-4- yljcarbamoylcyclopropanesulfonamide
Figure imgf000075_0002
This compound was obtained in 50% yield (580 mg) from 1.62g (4.52 mmol) of 1 -(3,5-dimethylisoxazol-4-yl)carbamoylcyclo-propanesulfonamide tert- butylcarbamate using 30 mL (120 mmol) of 4N HCl/dioxanes, stirring overnight, concentration and chromatography over a Biotage 4OM column (eluting with 0% to 5% methanol/dichloromethane: 1H NMR (methanol-dj) δ 1.57 (m, 2H), 1.61 (m 2H), 2.15 (s, 3H), 2.30 (s, 3H), 4.84 (s, 3H); 13C NMR (methanol-dO δ 9.65, 10.94, 15.01, 46.11, 114.82, 159.45, 165.55, 168.15; LRMS m/z 260 (M++H). Example 18 Preparation ofcyclobutylsulfonamidefrom cylobutyϊbromide
Figure imgf000075_0003
To a solution of 5.0 g (37.0 mmol) of cyclobutyl bromide in 30 mL of anhydrous diethyl ether (Et2O) cooled to -78 0C was added 44 mL (74.8 mmol) of 1.7M tert-butyl lithium in pentanes and the solution slowly warmed to —35 0C over 1.5h. This mixture was cannulated slowly into a solution of 5.0 g (37.0 mmol) of freshly distilled sulfuryl chloride in 100 mL of hexanes cooled to -400C, warmed to 0 0C over Ih and carefully concentrated in vacuo. This mixture was redissolved in Et2O, washed once with some ice-cold water, dried (MgSO4) and concentrated carefully. This mixture was redissolved in 20 mL of THF, added drop wise to 500 mL of saturated NH3 in THF and was allowed to stir overnite. The mixture was concentrated in vacuo to a crude yellow solid and was recrystallized from the minimum amount OfCH2Cl2 in hexanes with 1-2 drops of MeOH to afford 1.90 g (38%) of cyclobutylsulfonamide as a white solid. 1H NMR (CDCl3) δ 1.95-2.06 (m, 2H), 2.30-2.54 (m, 4H), 3.86 (p, J=8 Hz, IH), 4.75 (brs, 2H); 13C NMR (CDCl3) δ 16.43, 23.93, 56.29. HRMS m/z (M-H)- calcd for C4H8NSO2: 134.0276, found 134.0282. Example 19 Preparation of cyclopentyl sulfonamide
Figure imgf000076_0001
A solution of 18.5 mL (37.0 mmol) of 2M cyclopentyl-magnesium chloride in ether was added dropwise to a solution of 3.0 mL (37.0 mmol) freshly distilled sulfuryl chloride (obtained from Aldrich) in 100 mL of hexanes cooled to -78 0C. The mixture was warmed to 00C over 1 h and was then carefully concentrated in vacuo. This mixture was redissolved in Et2O (200 mL), washed once with some ice- cold water (200 mL), dried (MgSO4) and concentrated carefully. This mixture was redissolved in 35 mL of THF, added dropwise to 500 mL of saturated NH3 in THF and was allowed to stir overnite. The mixture was concentrated in vacuo to a crude yellow solid, the residue filtered through 5Og of silica gel using 70% EtOAc-hexanes as the eluent and the solution was then concentrated. The residue was recrystallized from the minimum amount OfCH2Cl2 in hexanes with 1-2 drops of MeOH to afford 2.49 g (41%) of cyclopentylsulfonamide as a white solid. 1H NMR (CDCl3) δ 1.58- 1.72 (m, 2H), 1.74-1.88 (m3 2H)5 1.94-2.14 (m, 4H), 3.48-3.59 (m, IH), 4.80 (bs, 2H); 13C NMR (CDCl3) 525.90, 28.33, 63.54; MS m/e 148 (M-H)". Example 20
Preparation ofcyclohexyl sulfonamide
Figure imgf000077_0001
A solution of 18.5 mL (37.0 mmol) of 2M cyclohexylmagnesium chloride (TCI Americas) in ether was added dropwise to a solution of 3.0 mL (37.0 mmol) freshly distilled sulfuryl chloride in 100 mL of hexanes cooled to —78 0C. The mixture was warmed to 0 0C over 1 h and was then carefully concentrated in vacuo. This mixture was redissolved in Et2O (200 mL), washed once with some ice-cold water (200 mL), dried (MgSO4) and concentrated carefully. This mixture was redissolved in 35 mL of THF, added dropwise to 500 mL of saturated NH3 in THF and was allowed to stir overnite. The mixture was concentrated in vacuo to a crude yellow solid, the residue filtered through 50g of silica gel using 70% EtOAc-hexanes as the eluent and was concentrated. The residue was recrystallized from the minimum amount of CH2CI2 in hexanes with 1-2 drops of MeOH to afford 1.66 g (30%) of cyclohexyl-sulfonamide as a white solid: 1H NMR (CDCl3) δ 1.11 -1.37 (m, 3H), 1.43-1.56 (m, 2H), 1.67-1.76 (m, IH), 1.86-1.96 (m, 2H), 2.18-2.28 (m, 2H), 2.91 (tt, J=12? 3.5.Hz, IH), 4.70 (bs, 2H); 13C NMR (CDCl3) δ 25.04, 25.04, 26.56, 62.74; MS m/e 162 (M-I)". Example 21 Preparation of Neopentylsulfonamide
Figure imgf000077_0002
Following the procedure for the preparation of cyclohexylsulfonamide. 49 mL (37 mmol) of 0.75M neopentylmagnesium chloride (Alfa) in diethyl ether was converted to 1.52g (27%) of neopentylsulfonamide as a white solid. 1H NMR (CDCl3) δ 1.17 (s, 9H), 3.12 (s, 2H), 4.74 (bra, 2H); 13C NMR (CDCl3) 529.46, 31.51, 67.38; MS m/e 150 (M-I)". Example 22
Preparation ofcyclobutylcarbinylsulfonamide
Figure imgf000078_0002
A solution of 12.3 g (83 mmol) of cyclobutylcarbinyl bromide (Aldrich) and 13.7g (91 mmol) of sodium iodide in 150 mL of acetone was refluxed overnight and then cooled to room temperature. The inorganic solids were filtered off and the acetone and cyclopropylcarbinyl iodide (8.41 g, 46%) distilled off at ambient and 150 torr at 80 0C, respectively.
A solution of 4.0 g (21.98 mmol) of cyclobutyl carbinyl iodide in 30 mL of anhydrous diethyl ether (diethyl ether) cooled to —78 0C was cannulated into a solution of 17 mL (21.98 mmol) of 1.3M sec-butyl lithium in cyclohexanes and the solution was stirred for 5 minutes. To this mixture was cannulated a solution of 3.0 g (21.98 mmol) of freshly distilled sulfuryl chloride in 110 mL of hexanes cooled to - 78 0C, the mixture warmed to room temperature over 1 hour and was then carefully concentrated in vacuo. This mixture was redissolved in diethyl ether, washed once with some ice-cold water, dried (MgSO^5 filtered, and concentrated carefully. This mixture was redissolved in 30 mL of THF, added dropwise to 500 mL of saturated NH3 in THF and was allowed to stir overnight. The mixture was concentrated in vacuo to a crude yellow solid and was recrystallized from the minimum amount of dichloromethane in hexanes with 1-2 drops of methanol to afford 1.39 g (42%) of cyclobutyl carbinylsulfonamide as a white solid. 1H NMR (CDCl3) δ 1.81-2.03 (m, 4H), 2.14-2.28 (m, 2H)5 2.81-2.92 (m, IH), 3.22 (d, J=I Hz, 2H), 4.74 (brs, 2H); 13C NMR (CDCl3) δ 19.10, 28.21, 30.64, 60.93; MS m/e 148 (M-I)". Example 23 Preparation ofcyclopropylcarbinylsulfonamide
Figure imgf000078_0001
Using the procedure employed for the preparation of cyclobutylcarbinylsulfonamide, cyclopropylcarbinylsulfonamide was prepared from cyclopropylcarbinyl bromide (Aldrich) (see also JACS 1981 , p.442-445). 1H NMR (CDCl3) δ 0.39-0.44 (m, 2H), 0.67-0.76 (m, 2H), 1.13-1.27 (m, IH), 3.03 (d, J=7.3 Hz, 2H), 4.74 (brs, 2H); 13C NMR (CDCl3) δ 4.33, 5.61, 59.93; MS m/e 134 (M-I). Example 24
Preparation ofcyclopropanesulfonic acid (l-(R)-amino-2-(S)-vinyl- cyclopropanecarbonyl)amide HCl salt
Figure imgf000079_0001
Step 1: Preparation of 1 (R)-tert-butoxycarbonylamino-2(S)-vinyl- cyclopropanecarboxylic acid
Figure imgf000079_0002
To a solution of l(R)-*er/-butoxycarbonylamino-2(S)-vinyl- cyclopropanecarboxylic acid ethyl ester (3.28 g, 13.2 mmol) in THF (7 mL) and methanol (7 mL) was added a suspension of LiOH (1.27 g, 53.0 mmol) in water (14 mL). The mixture was stirred overnight at room temperature and quenched with IN NaOH (15 mL) and water (20 mL). The resulting mixture was washed with ethyl acetate (20 mL), and the organic phase was extracted with 20 mL 0.5N NaOH. The combined aqueous phases were acidified with IN HCl until pH 4 and extracted with ethyl acetate (3 x 4OmL). The combined organic extracts were washed with brine, dried (MgSO4), filtered and concentrated to yield the title compound as a white solid (2.62 g, 87%). 1H NMR: (DMSO-de) ξ> 1.22-1.26 (m, IH), 1.37 (s, 9H), 1.50-1.52 (m, IH), 2.05 (q, J=9 Hz, IH), 5.04 (d, J=IO Hz, IH), 5.22 (d, J=17 Hz, IH), 5.64- 5.71 (m, 1 H), 7.18, 7.53 (s, NH (rotamers), 12.4 (br s, 1 H) ); MS m/z 228 (M++H). Step 2: Preparation ofcyclopropanesulfonic acid (1 -(R)-tert-butoxycarbonylamino- 2-(S)-vinylcyclopropanecarbonyl)-amide
Figure imgf000079_0003
A solution of the product of Step 1 (2.62 g, 11.5 mmol) and CDI (2.43 g, 15.0 mmol) in THF (40 mL) was heated at reflux for 50 minutes under nitrogen. The solution was cooled to room temperature and transferred by cannula to a solution of cyclopropylsulfonamide (1.82 g, 15.0 mmol) in THF (10 mL). To the resulting solution was added DBU (2.40 mL, 16.1 mmol) and stirring was continued for 20 hours. The mixture was quenched with IN HCl to pH 1 and THF was concentrated in vacuo. The suspension was extracted with ethyl acetate (2 x 50 mL) and the combined organic extracts were dried (Na2SO4), filtered, and concentrated. Purification by recystallization from hexanes-ethyl acetate (1 : 1) afforded the title compound (2.4 g) as a white solid. The mother liquor was purified by a Biotage 40S column (eluted 9% acetone in dichloromethane) to give a second batch of the title compound (1.1 g). Both batches were combined (total yield 92%). 1H NMR (DMSOd6) δ 0.96-1.10 (m, 4H), 1.22 (dd, /=5.5, 9.5 Hz, IH), 1.39 (s, 9H), 1.70 (t, /=5.5 Hz, IH)5 2.19-2.24 (m, IH), 2.90 (m, IH), 5.08 (d, /=10 Hz, IH), 5.23 (d, /=17 Hz, IH), 5.45 (m, IH), 6.85, 7.22 (s, NH (rotamers); MS m/z 331 (M++H). Step 3: Preparation of cyclopropanesulfonic acid (I-(R)-amino-2-(S)-vinyl- cyclopropanecarbonyl) amide HCl salt
Figure imgf000080_0001
A solution of the product of Step 2 (3.5 g, 10.6 mmol) in dichloromethane (35 mL) and TFA (32 mL) was stirred at room temperature for 1.5 hours. The volatiles were removed in vacuo and the residue suspended in IN HCl in diethyl ether (20 mL) and concentrated in vacuo. This procedure was repeated once. The resulting mixture was triturated from pentane and filtered to give the title compound as a hygroscopic, off-white solid (2.60 g, 92%). 1H NMR: (DMSO-dβ) δ 1.01-1.15 (m, 4H), 1.69-1.73 (m, IH), 1.99-2.02 (m, IH), 2.38 (q, /=9 Hz, IH), 2.92-2.97 (m, IH), 5.20 (d, /=11 Hz, IH), 5.33 (d, /=17 Hz, IH), 5.52-5.59 (m, IH), 9.17 (br s, 3H); MS m/z 231 (M+-HH). Example 25
Preparation of(JS,4R,6S,J4S,18R)-7-cis-J4-tert-butoxycarbonylamino-18-hydroxy- 2, 15-dioxo-3, 16-diazatricyclo[14.3.0.046]nonadec- 7-ene-4-carboxylic acid
Figure imgf000081_0001
Example 25 Step 1: Preparation ofl-(2(S)-tert-Butoxycarbonylamino-non-8-enoyl)-4(R)- hydroxy-pyrrolidine-2(S)-carboxylic acid methyl ester
Figure imgf000081_0002
A solution of 2(iS)-tert-butoxycarbonylamino-8-nonenoic acid (purchased from RSP Amino Acids)(3.5 g, 12.9 mmol) in 200 mL of DCM was treated sequentially with 4(/?)-hydroxypyrrolidine-2(5)-carboxylic acid methyl ester hydrochloride (2.15 g, 11.8 mmol), iV-methyl morpholine (4.25 mL, 38.6 mmol), and HATU (5.37 g, 14.1 mmol). The reaction mixture was stirred at rt under N2 for 3 days, and then concentrated in vacuo. The residue was partitioned between ethyl acetate and pH 4 buffer (biphthalate). The organic phase was washed with sat. aq. NaHCO3, dried (MgSO4), and concentrated in vacuo to give the crude product. Flash chromatography (50% ethyl acetate/hexane to 100% ethyl acetate) gave 4.7 g (~100%) of 1 -(2(5)-tert-Butoxycarbonylamino-non-8-enoyl)-4(i?)-hydroxy- pyrrolidine-2(<S)-carboxylic acid methyl ester as a colorless oil: 1H NMR (500 MHz, CD3OD) 5 1.33-1.50(m, 8 H), 1.46 (s, 9 H), 1.57 (m, 1 H), 1.72 (m, 1 H) 2.08 (m, 2 H)5 2.28 (m, 1 H), 3.72 (s, 3 H,) 3.75-3.87 (m, 2 H), 4.36 (m, 1 H), 4.51 (bs, 1 H), 4.57 (t, J=8.2 Hz, 1 H)5 4.95 (d, ./=10.4 Hz, 1 H), 5.01 (m, 1 H), 5.83 (m, 1 H); MS m/z 399 (M++l).
Step 2: Preparation ofl-{[l-(2(S)-tert-Butoxycarbonylamino-non-8-enoyl)-4(R)- hydroxy-pyrrolidine-2(S)carbonyl]-(lR)-amino}-2(S)-vinyl-cyclopropanecarboxylic acid ethyl ester '
Figure imgf000082_0001
l-(2(iS)-tert-Butoxycarbonylamino-non-8-enoyl)-4(i?)-hydroxy-pyrrolidine- 2(S)-carboxylic acid methyl ester (4.7 g, 11.8 mmol) was dissolved in THF (80 mL), methanol (20 mL), and water (40 mL). Powdered lithium hydroxide(5.6 g, 233 mmol) was added. The light yellow slurry was stirred at rt under N2 for 16 h, and then concentrated in vacuo. The residue was pardoned between ether and water. The ether phase was discarded, and the aqueous phase was treated with IN HCl until the pH was 4. This acidic solution was extracted with EtOAc (3x). The combined EtOAc extracts were dried (MgSO4) and concentrated in vacuo to give 4.36 g (96%) of l-(2(5)-/er?-butoxycarbonylamino-8-nonenoyl)-4(i?)-hydroxy-pyrrolidine-2(S)- carboxylic acid as a white solid. This acid was then dissolved in 150 mL of DMF and (li?,2,S^-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride (2.61 g, 13.6 mmol), iV-methyl morpholine (2.5 mL, 22.6 mmol), and HATU (5.2 g, 13.7 mmol) was added. The reaction mixture was stirred at rt under N2 for 16 h, and then concentrated in vacuo. The residue was partitioned between ethyl acetate and pH 4 buffer (biphthalate). The organic phase was washed with sat. aq. NaHCO3, dried (MgSO4), and concentrated in vacuo to give the crude product. Flash chromatography (60%-80% ethyl acetate/hexane) gave 6.0 g (98%) of 1-{[1-(2(S)- ^eri'-Butoxycarbonylamino-non-8-enoyl)-4(/?)-hydroxy-pyrrolidine-2(5)carbonyl]- (li?)-amino}-2(5)-vinyl-cycIopropanecarboxylic acid ethyl ester as a white solid: 1H NMR (500 MHz, CD3OD) 6 1.25 (t, J=7.2 Hz, 3 H), 1.33-1.80 (m, 10 H), 1.46 (s, 9 H), 2.09 (m, 3 H), 2.25 (m, 2 H), 3.76 (m, 2 H), 4.14 (m, 2 H), 4.27 (dd, J=8.5, 5.2 Hz, 1 H)5 4.50 (m, 2 H), 4.94 (d, J=10.1 Hz, 1 H), 5.01 (dd, J=17.1, 1.8 Hz, 1 H), 5.11 (dd, J=10.4, 1.8 Hz, 1 H), 5.30 (d, J=15.6 Hz, 1 H), 5.80 (m, 2 H), 8.57 (s, 1 H); MS m/z 522 (M++l).
Step 3: Preparation of(lS,4R,6S,14S,18R)-7-cis-14-tert-butoxycarbonylamino- 18-hydroxy-2, 15-dioxo-3, 16-diazatricyclo[l 4.3.0.04>6]nonadec- 7-ene-4-carboxylic acid ethyl ester
Figure imgf000083_0001
A solution of l-{[l-(2(5)-/erf-Butoxycarbonyl-amino-non-8-enoyl)-4(i?)- hy(koxy-pyτrolidirie-2(iS)carbonyl]-(li?)-amino}-2(5)-vinylcyclopropane-carboxylic acid ethyl ester (800 mg, 1.53 mmol) in 2 L of methylene chloride was flushed with N2 for 0.5 h. Then tricyclohexylphosphine[l,3-bis(254,6-trimethyl-phenyl)-4,5- dihydroimidazol-2-ylidene][benzylidene]-ruthenium (IV) dichloride (Strem) (64 mg, 0.075 mmol) was added, and the mixture was flushed with N2 for another 10 min. The light orange homogeneous solution was refluxed for 2 h to give a dark orange solution. The reaction mixture was cooled to rt and concentrated in vacuo to give an orange oil. Flash chromatography (ethyl acetate) gave 460 mg (61%) of (1 S,4R,6S, 145, 1 SR)-7-cis- 14-tert-butoxycarbonylamino- 18-hydroxy-2, 15-dioxo-
3,16-diazatricyclo[14.3.0.04'6]-nonadec-7-ene-4-carboxylic acid ethyl ester as a gray solid. 1H NMR (500 MHz, CDCl3) 5 1.19 (t, J=7.2 Hz, 3 H), 1.42 (s, 9 H), 1.22-1.8 (m, 8 H), 1.87 (m, 2 H), 2.03-2.22 (m, 4 H), 2.63 (m, 1 H), 3.65 (m, 1 H), 4.09 (m, 3 H)5 4.45 (m, 1 H), 4.56 (s, 1 H), 4.82 (m, 1 H), 5.23 (m, 1 H), 5.51 (s, 1 H), 7.16 (s, 1 H); MS m/z 494 (M++!).
Step 4: (IS, 4R, 6SJ4S, 18R)-7-cis-14-tert-butoxycarbonylamino-18-hydroxy-2, 15- dioxo-3, 16-diazatricyclofI 4.3.0.04' 6]-nonadec- 7-ene-4-carboxylic acid
Figure imgf000083_0002
To a solution of (l»5r,4i?565r,145, 18i?)-7-c/s-14-/ert-butoxycarbonylammo-18- hydroxy-2,15-dioxo-3, 16-diazatricyclo[l 4.3.0.04'6]-nonadec-7-ene-4-carboxylic acid ethyl ester (493 mg, 1.0 mmol) in THF (4 mL), methanol (1 mL), and water (2 mL), was added powdered lithium hydxoxide(480 mg, 20 mmol), and the light yellow slurry stirred at rt under N2 for 16 h. The mixture was then concentrated in vacuo and the residue partioned between ether and water. The ether phase was discarded, and the aqueous phase was treated with 1 N HCl until pH 4. This acidic solution was extracted with EtOAc three times. The combined EtOAc extracts were dried (MgSO4) and concentrated in vacuo to give 460 mg (98%) of Example 18, (1 S,4R,6S, 14S, 1 SR)-I -cis- 14-tert-butoxycarbonylamino- 18-hydroxy-2, 15-dioxo- 3,16-diazatricyclo[14.3.0.04'6]-nonadec-7-ene-4-carboxylic acid as a gray solid. 1H NMR (500 MHz, CD3OD) δ ppm 1.26 (t, J=7.2 Hz5 3 H), 1.35-1.52 (m, 15 H), 1.57- 1.68 (m, 3 H), 1.79 (m, 1 H), 2.04 (m, 1 H), 2.16-2.41 (m, 3 H), 3.80 (dd, J=10.7, 4.3 Hz, 1 H), 3.88 (m, 1 H), 4.38 (dd, J=8.9, 3.1 Hz, 1 H), 4.55 (m, 2 H)5 5.39 (t, J=9.8 Hz, 1 H), 5.58 (m, 1 H); MS m/z 466 (M++l). Example 26
Preparation of ft-Cyclopropanesulfonylaminocarbonyl-lS-hydroxy-l, 15-dioxo- 3,16-diaza-tricyclo[14.3.0.04'6]nonadec-7-en-14-yl)-carbamic acid tert-butyϊ ester
Figure imgf000084_0001
Step 1: Preparation ofl-{[l-(2-tert-Butoxycarbonylamino-non-8-enoyl)-4-(tert- butyl-dimethyl-silanyloxy)-pyrrolidine-2-carbonyl]-amino}-2- vinylcyclopropanecarboxylic acid ethyl ester
Figure imgf000085_0001
To a mixture of 1 - {[1 -(2(5)-/ert-Butoxycaxbonylamino-non-8-enoyl)-4(i?)- hydroxy-pyrrolidine-2(<S)carbonyl]-(l/?)-amino}-2(5)-vinyl-cyclopropanecarboxylic acid ethyl ester (1.5 g, 2.87 mmoL)in 10 mL of DMF was added imidazole (0.25 g, 3.67 mmoL) and tert-butyl-dimethylsilyl chloride (516 mg, 3.44 mmoL). The mixture was stirred at rt for two days. The reaction mixture was then concentrated in vacuo, and the residue was dissolved in ethyl acetate. This solution was washed with water, dried over magnesium sulfate, and concentrated in vacuo to obtain a crude solid. Purification by flash chromatography (eluting with 20% ethyl acetate in hexane) gave 1.43 g (78%) of l-{[l-(2-tert-butoxycarbonylamino-non-8-enoyl)-4- (tert-butyl-dimethyl-silanyloxy)-pyrrolidine-2-carbonyl] -amino} -2- vinylcyclopropanecarboxylic acid ethyl ester as a white solid. 1H NMR (300 MHz, CD3OD) δ 0.10 (s,6 H), 0.89 (s5 9 H)5 1.22 (m, 3 H), 1.31-1.48 (m, 16 H), 1.50-1.75 (m, 3 H), 2.06 (m, 3 H)5 2.11-2.33 (m, 2 H)5 3.70 (m, 2 H)5 4.03-4.19 (m, 2 H)5 4.21 (m, 1 H), 4.45 (t, J=7.87 Hz5 1 H)5 4.59 (m, 1 H), 4.91 (d, J=9.15 Hz5 1 H), 4.98 (d, J=I 7.20 Hz5 1 H)5 5.08 (dd5 J=I 0.25, 1.83 Hz, 1 H), 5.27 (dd, J=I 7.38, 1.65 Hz, 1 H), 5.65-5.87 (m5 2 H); MS m/z 636 (M++l). Step 2: Preparation ofl4-tert-Butoxycarbonylamino-18-(tert-butyl-dimethyl- silanyloxy)-2,l 5-dioxo-3, 16-diaza-tricyclo[14.3.0.04'6]nonadec-7-ene-4-carboxylic acid, ethyl ester
Figure imgf000086_0001
To a solution of l-{[l-(2-tert-butoxycarbonylamino-non-8-enoyl)-4-(tert- butyl-dimethyl-silanyloxy)-pyrrolidine-2-carbonyl]-amino}-2-vinyl- cyclopropanecarboxylic acid ethyl ester (1.63 g, 2.56 mmoL) in 640 mL of methylene chloride was added 215 mg (0.26 mmoL) of tricyclohexylphosphine[l,3- bis(2,4,6-tri[benzylidene]ruthenium(IV)dichloride. The mixture was heated at reflux for 15 min. The residue was concentrated in vacuo, and then purified by flash chromatography eluting with 30 % ethyl acetate/hexane. To further decolorize the sample, the crude product was chromatographed a second time eluting with 50% ether in hexane to give 1.5 g (96%) of 14-tert-butoxycarbonylamino-18-(tert-butyl- dimethyl-silanyloxy)-2, 15 -dioxo-3, 16-diaza-tricyclo[ 14.3.0.04)6]nonadec-7-ene-4- carboxylic acid ethyl ester as a white solid. 1H NMR (500 MHz, CD3Cl) δ 0.06 (s, 3 H), 0.07 (s, 3 H), 0.86 (s, 9 H), 1.18-1.24 (m, 6 H), 1.34-1.64 (m, 14 H), 1.86-1.96 (m, 3 H), 2.02-2.09 (m, 1 H), 2.11-2.17 (m, 1 H), 2.19-2.28 (m, 1 H), 2.57-2.63 (m, 1 H), 3.50-3.54 (m, 1 H), 3.71 (dd, /=10.22, 6.26 Hz, 1 H), 4.06-4.17 (m, 2 H), 4.52- 4.58 (m, 2 H), 4.75 (d, J=8.55 Hz, 1 H), 5.21 (t, J=9.92 Hz, 1 H), 5.35 (d, J=7.63 Hz, 1 H), 5.45-5.50 (m, 1 H), 6.94 (s,l H); MS m/z 608 (M++l). Step 3: Preparation ofl4-tert-butoxycarbonylamino-18-(tert-butyl-dimethyl- silanyloxy)-2,l 5-dioxo-3, 16-diaza-tricyclo[l 4.3.0.04'6]nonadec- 7-ene-4-carboxylic acid
Figure imgf000087_0001
To a solution of 14-tert-butoxycarbonylamino-18-(tert-butyl-dimethyl- silanyloxy)-2,15-dioxo-3,16-diaza-tricyclo[14.3.0.04>6]nonadec-7-ene-4-carboxylic acid ethyl ester (1.5g, 2.47mmoL) in a mixed solvent system of THF (4 mL), methanol (1 mL), and water (2 mL), was added powdered lithium hydroxide monohydrate (1.0 g, 50 mmoL). The light yellow slurry was stirred at rt under N2 for 4 h. The mixture was then concentrated in vacuo, and the residue partioned between ether and water. The ether phase was discarded, and the aqueous phase was treated with 1 N HCl until reaching pH 4. This acidic solution was extracted with EtOAc (3x). The combined EtOAc extracts were dried (MgSO4), and concentrated in vacuo to give 1.2 g (84%) of 14-tert-butoxycarbonylamino-l 8-(tert-butyl-dimethyl- silanyloxy)-2,15-dioxo-3,16-diaza-tricyclo[14.3.0.04'6]nonadec-7-ene-4-carboxylic acid as an off-white solid. 1H NMR (300 MHz, CD3OD) 0.12 (s, 6 H), 0.89 (s, 9 H), 1.23-1.64 (m, 17 H), 1.70-1.87 (m, 1 H), 1.90-2.49 (m, 6 H), 3.70-3.80 (m,l H), 3.83- 3.90 (m, 1 H), 4.28-4.36 (m, 1 H), 4.47-4.55 (m, 1 H), 4.65 (s, 1 H), 5.30-5.39 (m, 1 H)5 5.53-5.62 (m, 1 H); MS m/z 580 (M++l). Step 4: Preparation of[18-(tert-butyl-dimethyl-silanyloxy)-4- cyclopropanesulfqnylam inocar bony 1-2, 15-dioxo-3, 16-dia∑a- tricyclo[14.3.0.04'6]nonadec-7-en-14-yl]-carbamic acid tert-butyl ester
Figure imgf000088_0001
14-tert-Butoxycarbonylamino-l 8-(tert-butyl-dimethyl-silanyloxy)-2, 15- dioxo-3,16-diaza-tricyclo[14.3.0.04'6]nonadec-7-ene-4-carboxylic acid (500 mg, 0.86 mraoL) was dissolved in 25 mL of THF and treated with CDI (180 mg, 1.12 mmoL). (Care was taken to avoid moisture by using oven dried glassware and maintaining a dry N2 atmosphere). After refluxing the reaction mixture for 2 h, it was cooled to rt and treated sequentially with cyclopropylsulfonamide (135 mg, 1.12 mmoL) and DBU (170 mg, 1.12 mmoL). The reaction mixture was stirred for 4 h at rt, and the THF was removed by rotary evaporation. The residue was partitioned between ethyl acetate and pH 4 buffer. The organic phase was dried (MgSO4) and concentrated in vacuo to give the crude product. It was then purified by flash chromatography
(eluting with 33% ethyl acetate in hexane) to give 300 mg (51%) of [18-(tert-butyl- dimethyl-silanyloxy)-4-cyclopropanesulfonylaminocarbonyl-2, 15-dioxo-3, 16-diaza- tricyclo[14.3.0.04'6]nonadec-7-en-14-yl]-carbamic acid tert-butyl ester as a white solid. 1H NMR (300 MHz5 CD3OD) δ IH 0.07 (s, 3 H), 0.08 (s, 3 H), 0.85 (s, 9 H), 0.87-1.49 (m, 21 H), 1.73-1.95 (m, 3 H), 2.08-2.16 (m, 1 H), 2.25-2.36 (m, 2 H), 2.42-2.56 (m, 1 H), 2.85-2.93 (m, 1 H), 3.65-3.74(dd, J=10.61, 3.66 Hz, 1 H), 3.89 (d, J=I 0.25 Hz, 1 H), 4.34 (m, J=9.70, 9.70 Hz, 1 H), 4.43 (t, J=7.87 Hz, 1 H), 4.57 (s, 1 H), 4.94-5.01 (m, 1 H), 5.10 (d, J=8.78 Hz, 1 H), 5.66-5.75 (m, 1 H), 6.55 (s, 1 H), 10.13 (s, 1 H); MS m/∑ 683 (M++l). Step 5: Preparation of (4-Cyclopropanesulfonylaminocarbonyl-l8-hydroxy-2,15- dioxo-3, 16-diaza-tricyclo[l 4.3.0. G4'6] nonadec- 7-en-14-yl)-carbamic acid tert-butyl ester
Figure imgf000089_0001
To a mixture of [ 18-(tert-butyl-dimethylsilanyloxy)-4- cyclopropanesulfonylaminocarbonyl-2,15-dioxo-3, 16-diaza- tricyclo[14.3.0.04>6]nonadec-7-en-14-yl]-carbamic acid tert-butyl ester (33Omg, 0.48 mmoL) in 25 mL of THF was added tetrabutylammonium floride (150 mg, 0.54 mmoL). The reaction mixture was stirred at rt for 18 h, and then the THF was removed by rotary evaporation. The residue was partitioned between ethyl acetate and water. The organic phase was dried (MgSO4) and concentrated in vacuo to give the crude product. It was then purified by triturating with hexane to yield 200 mg (73%) of (4-cyclopropanesulfonylaminocarbonyl- 18-hydroxy-2, 15-dioxo-3, 16-diaza- tricyclo[14.3.0.04>6]nonadec-7-en-14-yl)-carbamic acid tert-butyl ester, Example 19, as a white solid. 1H NMR (500 MHz, CD3Cl) δ 1.87-1.64 (m, 21 H), 1.70-1.98 (m, 3 H), 2.15-2.56 (m, 5 H), 2.85-2.94 (m, 1 H), 3.71 (d, J=13.91 Hz, 1 H), 4.10-4.26 (m, 2 H), 4.51 (t, J=7.87 Hz, 1 H), 4.62 (s, 1 H), 4.98 (m, 1 H), 5.06 (d, J=8.78 Hz, 1 H), 5.64-5.71 (m, 1 H), 6.72 (s, 1 H), 10.24 (s, 1 H); MS m/z 569 (M++l).
The following macrocyclic alcohol intermediates A and B were prepared employing the procedures described in examples 25 and 26:
Figure imgf000089_0002
The following macrocyclic alcohol intermediates C, D, E, F could be prepared employing the chemistry described and referenced herein as for example in Examples
25 and 26:
Figure imgf000090_0001
Example 27
Preparation of Example 27, 2(S)-tert-butoxycarbonylamino-3-pent-4- enylsulfanylpropionic acid
Figure imgf000090_0002
Step 1: To a solution of 7V-Boc-cysteine methyl ester (3.36 g, 0.014 mol) in methanol (166 mL) at RT was added triethylamine (10.8 mL) and l-bromopent-4-ene (3.19 g, 21 mmol, 1.5 equivalents) and the resulting solution was stirred at room temperature overnight. The mixture was then concentrated in vacuo and the resulting residual mixture was purified using flash chromatography (hexane, ethyl acetate gradient) to provide 1.76 g (41%) of the desired thioether. 1H NMR (500 MHz, CDCl3) δ 1.43 (s, 9H), 1.64 (m, 2H), 2.11 (m, 2H), 2.51 (m, 2H), 2.95 (m, 2H), 3.75 (s, 3H), 4.51 (m, IH), 4.95-5.03 (m, 2H), 5.34 (m, IH), 5.80 (IH, m); MS m/z 304(M++l). Step 2: The thioether product of step 1 (9.51 g, 31.4 mmol) was added to a mixture of IM LiOH in water (20OmL) and THF (20OmL) and the resulting mixture was stirred at room temperature overnight. The reaction mixture was then acidified using IN hydrochloric acid and the resulting mixture was extracted several times with ethyl acetate. The extracts were combined, dried over magnesium sulfate, and concentrated in vacuo to provide the desired acid, Example 27, which was used as is in the next reaction. Example 28 Preparation of Example 28, N-tert-Butoxycarbonyl-3-(4-pentenylthio)-L-valine
Figure imgf000091_0001
Example 28
Step 1: Preparation of N-tert-butoxycarbonyl-3-(4-pentenylthio)-L-valine, methyl ester
Figure imgf000091_0002
To a solution of 7.12g (48 mmol, 1.0 eq) of L-penicillamine in 10OmL of 1,4- dioxane and 25mL of water at room temperature was added 9.6OmL (96 mmol, 2.0 eq) of ION aqueous sodium hydroxide solution, followed by the dropwise addition of 12.0OmL (101 mmol, 2.1 eq) of 5-bromo-l-pentene over several minutes. The resulting mixture was stirred at room temperature for 68 hours. At this point 12.5Og (57 mmol, 1.2 eq) of di-tert-butyl dicarbonate was added, and the mixture was stirred at room temperature for another 6 hours. The mixture was concentrated under vacuum, and the residue was dissolved in water. The aqueous mixture was washed with diethyl ether, adjusted to pH 3 employing IN hydrochloric acid, and then extracted with ethyl acetate. The combined extracts were washed with brine, dried over anhydrous magnesium sulfate, filtered, and concentrated under vacuum. The crude product (12.2Og) was dissolved in 12OmL of anhydrous dimethylsulfoxide. To this solution was added 10.50g (76 mmol) of potassium carbonate and 4.7OmL (76 mmol) of iodomethane, and the resulting mixture was stirred at room temperature for 24 hours. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined extracts were washed with water (2X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. Column chromatography on silica gel (elution: 2 -10% ethyl acetate/hexane) provided 8.54g of N-tert-butoxycarbonyl-3-(4-pentenylthio)-L- valine, methyl ester as a colorless oil. NMR (300 MHz, CDCl3): δ 5.76 (d of d oft, 1 H, J = 17.2, 10.3, 6.6 Hz), 5.35 (br d, 1 H, J = 9.0 Hz), 5.05-4.94 (m, 2 H), 4.27 (br d, 1 H5 J = 9.0 Hz), 3.73 (s, 3 H), 2.52 (m, 2 H), 2.13 (quart., 2 H5 J = 7.3 Hz), 1.61 (quint., 2 H, J = 7.3 Hz), 1.43 (s, 9 H), 1.35 (s, 3 H), 1.33 (s, 3 H). Step 2: Preparation of Example 28, N-tert-Butoxycarbonyl-3-(4-pentenylthio)-L- valine
Figure imgf000092_0001
Example 28
To a solution of 8.52g (25.7 mmol) of N-tert-butoxycarbonyl-3-(4- pentenylthio)-L- valine, methyl ester in 20OmL of tetrahydrofuran at room temperature was added a solution of 1.1 Og (26.2 mmol) of lithium hydroxide monohydrate in 5OmL of water. The resulting mixture was stirred at room temperature for 65 hours. To the reaction mixture then was added 28mL of 1.00N hydrochloric acid. The mixture was diluted with diethyl ether, washed with water (3X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to afford 8.10g of N-tert-butoxycarbonyl-3-(4-pentenylthio)-L-valine as a colorless oil. NMR (300 MHz, CDCl3): δ 5.75 (d of d oft, 1 H, J = 17.2, 10.3, 6.6 Hz), 5.40 (br s, 1 H), 5.05-4.94 (m, 2 H), 4.28 (br s, 1 H), 2.56 (m, 2 H), 2.13 (quart., 2 H, J = 7.3 Hz), 1.63 (quint, 2 H, J = 7.3 Hz), 1.44 (s, 9 H), 1.39 (s, 3 H), 1.37 (s, 3 H). Example 29
Preparation of Example 29, 5-Allyloxy-2(S)-(tert-butoxycarbonylamino)pentanoic acid
Figure imgf000093_0001
Step 1: Preparation of Isopropyl pyrrolidin-5-one-2(S)-carboxylate
Figure imgf000093_0002
A solution of L-pyro glutamic acid (Aldrich, 25.0 g, 195 mmol) and para- toluenesulfonic acid mono hydrate (3.71 g, 19.5 mmol) was refluxed in isopropanol (40 mL) under nitrogen for 6 hours using a Dean-Stark trap variation (condensate returned through a Soxhlet extractor filled with 4 A molecular sieves). After cooling to room temperature, the reaction was diluted with ether, washed with saturated aqueous sodium bicarbonate and then saturated aqueous NaCl, dried (MgSO4) and evaporated to give a colorless syrup. It crystallized upon setting. Triturating the crystalline residue in hexane provided 31.9 g (96%) of isopropyl pyrrolidin-5-one- 2(S)-carboxylate as white prisms: 1H NMR (300 MHz, Chloroform-D) δ 6.35 (br s, 1 H), 5.04 (sept. I H1 J = 6.2 Hz), 4.18 (dd, I H1 J= 8.4, 5.3 Hz), 2.51-2.28 (m, 3 H), 2.27-2.12 (m, 1 H), 1.24 (d, 6 H, J = 6.2 Hz). LCMS m/z 172 (M-HH)+. Step 2: Preparation of Isopropyl l-(tert-butoxycarbonyl)-pyrrolidin-5-one-2(S)~ carboxylate
Figure imgf000093_0003
A solution of isopropyl pyrrolidin-5-one-2(S)-carboxylate (product of step 26A5 31.9 g, 188 mmol), di-tert-butyl dicarbonate (48.6 g, 225 mmol) and DMAP (2.30 g, 8.8 mmol) in acetonitrile (300 mL) was stirred at room temperature under N2 for 30 minutes. The reaction was evaporated to about 100 mL, diluted with ether, washed with IN HCl then saturated aqueous NaCl, dried (MgSO4) and evaporated to give isopropyl l-(tert-butoxycarbonyl)pyrrolidin-5-one-2(S) carboxylate as a light yellow oil, 50.1 g (99%): 1H NMR (300 MHz, Chloroform-D) δ 5.06 (sept. 1 H, J = 6.2 Hz), 4.53 (dd, 1 H, J = 9.5, 2.9 Hz), 2.66-2.40 (m, 2H), 2.36-2.22 (m, 1 H), 2.03- 1.93 (m, 1 H), 1.47 (s, 9 H), 1.26 (d, 3 H, J = 6.2 Hz), 1.24 (d, 3 H, J = 6.2 Hz). LCMS m/z 272 (M+H)+.
Step 3: Preparation of Isopropyl 2(S)-(tert-butoxycarbonylamino)-5- hydroxypentanoate
Figure imgf000094_0002
To a solution of isopropyl l-(tert-butoxycarbonyl)pyrrolidin-5-one-2(S)- carboxylate (product of step 26B, 49.5 g, 183 mmol) in methanol (300 mL) was added sodium borohydride (10.0 g, 263 mmol) in ~1 g portions over 1.5 hours. The reaction was stirred under nitrogen for another 10 minutes. It was diluted with water, extracted with ether, combined organic fractions washed with saturated aqueous NaCl, dried (MgSO4) and evaporated to give a light yellow oil. Flash chromatography (silica gel, 20-30% ethyl acetate/hexane) gave 31.8 g (64%) of isopropyl 2(S)-(tert-butoxycarbonylamino)-5-hydroxypentanoate as a colorless syrup: 1H NMR (300 MHz5 Chloroform-D) δ 5.16 (br d, 1 H, J = 7.3 Hz), 5.03 (sept., 1 H, J = 6.2 Hz), 4.28 (br d, 1 H, J = 6.2 Hz), 3.67 (br dd, J = 10.2, 5.5 Hz), 1.94-1.79 (m, 2 H), 1.76-1.67 (m, 1 H), 1.66-1.56 (m, 2 H), 1.43 (s, 9 H), 1.25 (d, 3 H, J = 6.2 Hz), 1.23 (d, 3 H, J = 6.2 Hz). LCMS m/z 276 (M+H)+. Step 4: Preparation ofIsopropyl-5-allyloxy-2(S)-(tert- butoxycarbonylamino)pentanoate
Figure imgf000094_0001
A degassed mixture of isopropyl 2(S)-(tert-butoxycarbonylamino)-5- hydroxypentanoate (product of step 26C, 17.6 g, 63.9 mmol), allyl methyl carbonate (24.0 ml, 213 mmol), Pd2(dba)3 (1.62 g, 1.78 mmol) and BINAP (4;42 g, 7.10 mmol) in THF (150 mL) was refluxed under nitrogen for 3 hours. After cooling to room temperature, the reaction was diluted with ether, filtered through celite and evaporated giving a dark brown syrup. Flash chromatography of the residue (silica gel, 30% ether/hexane) gave isopropyl 5-allyloxy-2(S)-(tert- butoxycarbonylamino)pentanoate as a viscous colorless oil, 16.3 g (81%): 1H NMR (300 MHz, Chloroform-D) δ 5.88 (ddt, 1 H5 17.4, 10.4, 5.5), 5.28 (m, 1 H)5 5.22-5.11 (m, 1 H), 5.02 (sept., 1 H, J = 6.2 Hz), 4.21 (br t, 1 H, J = 6.7 Hz), 3.94 (dt, 2 H5 J = 5.9, 1.5 Hz), 3.42 (t, 2 H, J = 5.9 Hz)5 1.90-1.82 (m, 1 H)5 1.75-1.57 (m, 3 H), 1.42 (s, 9 H), 1.21 (d, 3 H5 J = 6.2 Hz), 1.19 (d, 3 H5 J = 6.2 Hz). LCMS m/z 316 (M+H)+. Step 5: Preparation of 5-Allyloxy-2(S)-(tert-butoxycarbonylamino)pentanoic acid
Figure imgf000095_0001
A mixture of isoprop 5-allyloxy-2(S)-(tert- butoxycarbonylamino)pentanoate (product of step 26D5 16.1 g, 51.1 mmol) and lithium hydroxide hydrate (4.19 g, 102 mmol) in THF/water (100 mL/ 20 mL) was stirred at room temperature under nitrogen for 16 hours. The reaction was diluted with water, washed with ether, pH of aqueous fraction adjusted to ~4, extracted with ether, combined organic fractions washed with saturated NaCl, dried (MgSO4) and evaporated giving 5-allyloxy-2(S)-(tert-butoxycarbonylamino)pentanoic acid as a light yellow syrup: 1H NMR (300 MHz, Chloroform-D) δ 5.89 (ddt, 1 H5 J = 17.4, 10.4, 5.5), 5.25 (dd, 1 H, J = 17.4, 1.6 Hz)5 5.17 (dd5 I H1 J= 10.4, 1.6 Hz), 4.30 (br d, 1 H, J = 6.2), 3.96 (dt, 2 H, J = 5.9, 1.5 Hz), 3.46 (t, 2 H, J = 5.9 Hz), 1.96-1.86 (m, 1 H), 1.85-1.77 (m, 1 H), 1.75-1.64 (m, 2 H)5 1.43 (s, 9 H). LCMS m/z 274 (M+H)+. Example 30
General Procedure for the Preparation of Example 30
Figure imgf000096_0001
Example 23 was prepared by adding a DMF solution of N-trityl protected threonine to a DMF solution of sodium hydride cooled to — 15 C. The reaction mixture was stirred for 30 minutes at— 15 C after which 5-bromo-l-pentene was added and the resulting mixture was warmed to —5 C. The reaction mixture was maintained at —5 C for 3 days after which time the reaction was quenched by the addition of IN aqueous HCl and worked up using standard extraction procedures as described above. Example 23 was obtained in pure form by standard chromatography procedures. Example 31: Preparation of Example 31, N-tert-Butoxycarbonyl-O-(4-pentenyl)-L-serine
Figure imgf000096_0002
Example 31 Step 1: Preparation ofN-tert-Butoxycarbonyl-O-(4-pentenyl)-L-serine, methyl ester
Figure imgf000096_0003
To a solution of 10.26g (50 mmol, 1.0 eq) of N-tert-butoxycarbonyl-L-serine in 50OmL of anhydrous dimethylsulfoxide at room temperature was added 2.0Og (50 mmol, 1.0 eq) of 60% sodium hydride in mineral oil. This mixture was stirred at room temperature for 0.5 hour until the evolution of gas had ceased. To the resulting solution was added 6.0OmL (50 mmol, 1.0 eq) of 5-bromo-l-pentene followed immediately by another 2.0Og (50 mmol, 1.0 eq) of 60% sodium hydride in mineral oil. The reaction mixture then was stirred at room temperature for 16 hours. The mixture was diluted with 200OmL of water, adjusted to pH 3-4 by the addition of 5OmL of 1.00N hydrochloric acid, and extracted with ethyl acetate. The organic phase was washed with water (2X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. To remove the residual mineral oil the resulting material was dissolved in a dilute aqueous sodium hydroxide solution. This aqueous solution was washed with hexane and then adjusted to pH 4 employing hydrochloric acid, and extracted with ethyl acetate. The extract was washed with water (2X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. The crude product (7.7Og) was dissolved in 10OmL of anhydrous dimethylsulfoxide. To this solution was added 7.8Og (56 mmol) of potassium carbonate and 3.5OmL (56 mmol) of iodomethane, and the resulting mixture was stirred at room temperature for 24 hours. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined extracts were washed with water (2X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum. Column chromatography on silica gel (elution: 2 -10% ethyl acetate/hexane) provided 6.7Og of N-tert-butoxycarbonyl-O-(4-pentenyl)-L-serine, methyl ester as a colorless oil. NMR (300 MHz, CDCl3): δ 5.78 (d of d oft, 1 H, J = 17.2, 10.2, 6.6 Hz), 5.34 (br d, 1 H, J = 8.0 Hz), 5.03-4.92 (m, 2 H), 4.40 (m, 1 H), 3.81 (d of d, 1 H, J = 9.5, 2.9 Hz), 3.74 (s, 3 H), 3.61 (d of d, 1 H, J = 9.5, 3.5 Hz), 3.42 (m, 2 H), 2.06 (quart., 2 H, J = 7.3 Hz), 1.61 (quint., 2 H, J = 7.3 Hz), 1.44 (s, 9 H). Step 2: Preparation of Example 31, N-tert-Bιttoxycarbonyl-O-(4-pentenyl)-L-serine
Figure imgf000097_0001
Example 31 To a solution of 6.65g (23 mmol) of N-tert-butoxycarbonyl-0-(4-pentenyl)-L- serine, methyl ester in 50OmL of tetrahydrofuran at room temperature was added a solution of 1.95g (46 mmol) of lithium hydroxide monohydrate in 10OmL of Water. The resulting mixture was stirred at room temperature for 40 hours. To the reaction mixture then was added 46mL of 1.00N hydrochloric acid. The mixture was diluted with ethyl acetate, washed with water (3X) and brine, dried over anhydrous sodium sulfate, filtered, and concentrated under vacuum to afford 6.30g of N-tert- butoxycarbonyl-O-(4-pentenyl)-L-serine as a colorless oil. NMR (300 MHz, CDCI3): δ 5.77 (d of d oft, 1 H, J = 17.2, 10.2, 6.6 Hz), 5.37 (br d, 1 H, J = 8.0 Hz), 5.03-4.92 (m, 2 H), 4.42 (m, 1 H), 3.87 (d of d5 1 H, J = 9.5, 2.6 Hz), 3.63 (d of d, 1 H5 J = 9.5, 4.0 Hz), 3.45 (t, 2 H1 J = 6.6 Hz), 2.07 (quart., 2 H, J = 7.3 Hz), 1.64 (quint., 2 H, J = 7.3 Hz), 1.44 (s, 9 H). Example 32
Preparation of(S)-4-allyloxy-2-(tert-butoxycarbonylamino)butyric acid
Figure imgf000098_0001
To a mixture of sodium hydride (913 mg, 22.8 mmoL) in DMF at 00C was added N-t-Boc-L-homoserine (2 g, 9.13 mmoL). This reaction mixture was stirred at 00C for 15 min, and then allyl bromide (1.38 g, 11.4 mmoL) was added. The mixture was warmed up to rt, and stirred for 2 h. It was then concentrated in vacuo. The residue was diluted with water, and sequentially washed with hexane and ether. The organic layers were discarded, and the aqueous layer was carefully adjusted to pH 3 with 1 N HCl. This acidic aqueous solution was extracted with ethyl acetate. The organic phase was dried (MgSO-j), and concentrated in vacuo to yield 2.2 g (93%) of (S)-4-allyloxy-2-(ferM3utoxycarbonylamino)butyric acid as a colorless oil. 1H NMR (300 MHz, CD3OD) 6 1.42 (s, 9 H), 1.80-1.90 (m, 1 H), 2.04-2.16 (m, 1 H), 3.50- 3.54 (m, 2 H), 3.97 (d, J=4.39 Hz, 2 H), 4.23 (dd, J=8.78, 4.39 Hz, 1 H), 5.15 (d, J=10.25 Hz, 1 H), 5.26 (dd, J=17.38, 1.65 Hz, 1 H)5 5.84-5.97 (m, 1 H). Example 33
Figure imgf000099_0001
Compound 1
Step 1: Preparation ofl4-tert-butoxycarbonylamino-2,15-dioxo-18-(pyridin-2- yloxy)-3, 16-diaza-tricyclo[14.3.0.04'6]nonadec- 7-ene-4-carboxylic acid
Figure imgf000099_0002
To a mixture of (14-tert-butoxycarbonylamino-18-hydroxy-2,15-dioxo-3,16- diaza-tricyclo[14.3.0.04>6]nonadec-7-ene-4-carboxylic acid (150 mg, 0.32 mmol; prepared in Ex. 25, step 4) in 3 mL of DMSO was added potassium t-butoxide (91 mg, 0.81 mmol). The mixture was stirred at rt for 5 min, and then 2-fluoro-pyridine (39 mg, 0.40 mmoL) was added and the mixture was stirred at rt overnight. The reaction was quenched by adding 10 mL of water, and then the pH was adjusted to 4 using 0.1 N HCL. It was then extracted with ethyl acetate, dried, concentrated in vacuo to obtain a yellow oil. This crude product was purified by flash chromatography on silica gel eluting with 10 % methanol in ethyl acetate to isolate 100 mg (56%) of 14-tert-butoxycarbonylamino-2, 15 -dioxo- 18-(pyridin-2-yloxy)- 3,16-diaza-tricyclo[14.3.0.04'6]nonadec-7-ene-4-carboxylic acid as a white solid. 1H NMR (500 MHz, MeOD) δ 1.24 - 1.53 (m, 6 H), 1.36 (s, 9 H), 1.55 - 1.68 (m, 3 H), 1.73 - 1.90 (m, 1 H), 2.02 - 2.11 (m, 1 H), 2.16 (q, J=8.65 Hz, 1 H), 2.35 - 2.45 (m, 1 H), 2.51 (dd, J=7.63, 2.75 Hz, 2 H), 4.06 - 4.14 (m, 1 H), 4.27 (d, J=I 1.29 Hz, 1 H), 4.33 (s, 1 H)5 4.58 (t, J=7.93 Hz, 1 H), 5.46 - 5.57 (m, 2 H), 5.70 (s, 1 H), 6.79 (d, J=8.54 Hz, 1 H), 6.93 - 7.02 (m, 1 H), 7.68 (t, J=7.63 Hz, 1 H), 8.16 (d, J=4.58 Hz, 1 H). LC-MS (YMC Xterra ODS S7 micrometer column: 3.0x50 mm length. Gradient: 100% Solvent A/0% Solvent B to 0% Solvent A/ 100% Solvent B. Gradient time: 4 min. Hold time: 1 min. Flow rate: 4 mL/min. Detector Wavelength: 220 am. Solvent A: 10% MeOH / 90% H2O / 0.1% TFA. Solvent B: 10% H2O / 90% MeOH / 0.1% TFA.)(Retention time: 2.90 min), MS m/x 543 (M++l). Step 2: Preparation of compound 1
14-tert-Butoxycarbonylamino-2,15-dioxo-18-(pyridin-2-yloxy)-3,16-diaza- tricyclo[14.3.0.04'6]nonadec-7-ene-4-carboxylic acid (100 mg, 0.18 mmol) was dissolved in 5 mL of THF and treated with carbonyl diimidazole (39 mg, 0.24 mmol). (Care was taken to avoid moisture by using oven dried glassware and maintaining a dry N2 atmosphere). After refluxing the reaction mixture for one hour, it was cooled to rt and treated sequentially with cyclopropylsulfonamide (29 mg, 0.24 mmol) and DBU (36 mg, 0.24 mmol). After stirring for 24 h at rt, the THF was removed by rotary evaporation. The residue was partitioned between ethyl acetate and pH 4 buffer. The organic phase was dried (MgS 04) and concentrated in vacuo to give the crude product. The resulting oil was dissolved in methanol and purified by preparative HPLC ( YMC ODS-A, S5, 30x50 mm, gradient:40% B to 85% B, 8 min, hold 2 min, flow rate 25 mL/min) to isolate compound 1 as a white powder (30 mg, 26%). NMR (500 MHz, CDCl3) δ 0.89 - 0.97 (m, 1 H), 1.04 - 1.17 (m, 2 H), 1.23 - 1.36 (m, 6 H), 1.33 (s, 9 H), 1.44 - 1.51 (m, 3 H), 1.57-1.68 (m, 1 H), 1.83 - 1.96 (m, 2 H), 2.22 - 2.32 (m, 1 H), 2.45 - 2.61 (m, 3 H), 2.85 - 2.95 (m, 1 H), 3.90 - 4.00 (m, 1 H), 4.26 (d, J=I 1.29 Hz, 1 H), 4.32 (s, 1 H), 4.56 (s, 1 H), 4.94 - 5.01 (m, 1 H), 5.15 (d, J=6.41 Hz, 1 H), 5.63 - 5.77 (m, 2 H), 6.66 (d, J=8.24 Hz, 1 H), 6.72 (s, 1 H), 6.84 - 6.94 (m, 1 H), 7.56 (t, J=7.78 Hz, 1 H), 8.13 (d, J=4.27 Hz, 1 H), 10.18 (s, 1 H). LC-MS (YMC Xterra ODS S7 micrometer column: 3.0x50 mm length. Gradient: 100% Solvent A/0% Solvent B to 0% Solvent A/100% Solvent B. Gradient time: 4 min. Hold time: 1 min. Flow rate: 4 mL/min. Detector Wavelength: 220 nm. Solvent A: 10% MeOH / 90% H2O / 0.1% TFA. Solvent B: 10% H2O / 90% MeOH / 0.1% TFA.)(Retention time: 3.05 min), MS m/z 646 (M++l). Example 34
Figure imgf000101_0001
iSte/? i: Preparation ofl4-tert-Butoxycarbonylamino-18-(4-nitrophenoxy)-2,15- dioxo-3,16-diazatricyclo[14.3.0.0 ' Jnonadec-V-ene^-carboxylic acid
Figure imgf000101_0002
To a mixture of (14-tert-butoxycarbonylamino-18-hydroxy-2,15-dioxo-3,16- diaza-tricyclo[14.3.0.04'6]nonadec-7-ene-4-carboxylic acid, ethyl ester (192 mg, 0.39 mmol; prepared in Ex. 25, step 3) in 3 mL of THF was added sodium hydride (50 mg, 60% in oil, 1.25 mmol). The mixture was stirred at rt for 5 min. then l-fluoro-4- nitrobenzene(60 mg, 0.42 mmol) was added and stirring was continued at rt overnight. The reaction was quenched by adding 10 mL of water, and then 0.1 N hydrochloric acid was used bring the pH to 4. This acidic solution was then extracted with ethyl acetate. The organic phase was dried over magnesium sulfate and concentrated in vacuo to obtain 14-tert-butoxycarbonylamino-l 8-(4-nitrophenoxy)- 2,15-dioxo-3,16-diaza-tricyclo[14.3.0.04,6]nonadec-7-ene-4-carboxylic acid as a white solid (100 mg, 44%). LC-MS (YMC Xterra MS C18 S7 column: 3.0x50 mm length. Gradient: 100% Solvent A/0% Solvent B to 0% Solvent A/100% Solvent B. Gradient time: 4 min. Hold time: 1 min. Flow rate: 4 mL/min. Detector Wavelength: 220 nm. Solvent A: 10% MeOH / 90% H2O / 0.1% TFA. Solvent B: 10% H2O / 90% MeOH / 0.1% TFA.)(Retention time: 3. 17 min), MS m/z 587(M++1). Step 2: Preparation of Compound 2
14-tert-ButoxycarbonyIamino-18-(4-nitro-phenoxy)-2,15-dioxo-3,16-diaza- tricyclo[14.3.0.04,6]nonadec-7-ene-4-carboxylic acid (100 mg, 0.17 mmol) was dissolved in 5 mL of THF and treated with carbonyl diimidazole (38 mg, 0.23 mmol). (Care was taken to exclude moisture by using oven dried glassware and maintaining a dry N2 atmosphere). After refluxing the reaction mixture for one hour, it was cooled to rt and treated sequentially with cyclopropylsulfonamide (29 mg, 0.24 mmol) and DBU (36 mg, 0.24 mmoL). After stirring for 24 h at rt, the THF was removed by rotary evaporation. The residue was partitioned between ethyl acetate and pH 4 buffer. The organic phase was dried (MgSO4) and concentrated in vacuo to give the crude product. Flash chromatography (50% ethyl acetate in hexane) gave 30 mg
(26%) of compound 2. LC-MS (YMC Xterra MS Cl 8 S7 column: 3.0x50 mm length. Gradient: 100% Solvent A/0% Solvent B to 0% Solvent A/100% Solvent B. Gradient time: 4 min. Hold time: 1 min. Flow rate: 4 mL/min. Detector Wavelength: 220 nm. Solvent A: 10% MeOH / 90% H2O / 0.1% TFA. Solvent B: 10% H2O / 90% MeOH / 0.1% TFA.)(Retention time: 3. 21 min), MS m/z 690(M++l). Example 35
Figure imgf000102_0001
Preparation of Compound 3
To a mixture of (4-cyclopropanesulfonylaminocarbonyl-18-hydroxy-2,15- dioxo-3, 16-diaza-tricyclo[ 14.3.0.04'6]nonadec-7-en- 14-yl)carbamic acid tert-butyl ester (20 mg, 0.035 mmol; prepared in Ex. 26, step 5) in DMF (2 mL) was added t- BuOK (20 mg, 0.15 mmol) and l-chloro-6-iluoro-5-methoxyisoquinoline (15 mg, 0.07 mmol). The reaction was stirred for 16 h at rt. The reaction mixture then was partitioned between ether (10 mL) and water (5 mL). The aqueous phase was acidified to pH 4 using 1 N HCl. The resulting solution was extracted with EtOAc (3 x 20 mL). The combined EtOAc extracts were dried (MgSO4), filtered, and concentrated in vacuo to give a white solid. This crude product was purified by preparative HPLC (YMC Xterra, S5, 19 x 50mm, 60% to 100%B, gradient 15 min, hold 2 min, flow rate 25 mL/min ) to give 10 mg (38%) of the compound 3 as a white powder: LC-MS (YMC Xterra S7 column: 3.0x50 mm length. Gradient: 100% Solvent A/0% Solvent B to 0% Solvent A/100% Solvent B. Gradient time: 3 min. Hold time: 1 min. Flow rate: 4 mL/min. Detector Wavelength: 220 nm. Solvent A: 10% MeOH / 90% H2O / 0.1% TFA. Solvent B: 10% H2O / 90% MeOH / 0.1% TFA.)(Retention time: 2.53 min), MS m/z 760 (M++1).1H NMR (500 MHz, CDCl3) 6 ppm 0.89 - 0.97 (m, 1 H), 1.04 - 1.16 (m, 3 H), 1.20 - 1.51 (m, 7 H), 1.30 (s, 9 H), 1.54-1.64 (m, 1 H), 1.73-1.96 (m, 3 H), 2.22-2.31 (m, 1 H), 2.47-2.57 (m, 1 H), 2.60- 2.67 (m, 2 H), 2.86-2.94 (m, 1 H), 3.97 (s, 3 H), 3.99 - 4.10 (m, 1 H), 4.31 (t, J=7.63 Hz, 1 H), 4.45 (d, J=I 1.29 Hz, 1 H), 4.65 (t, J=7.48 Hz, 1 H), 4.97 (t, J=9.46 Hz, 1 H), 5.16 (d, J=7.93 Hz, 1 H), 5.32 (s, 1 H), 5.71 (q, J=8.95 Hz, 1 H), 6.84 (s, 1 H), 7.10 (s, 1 H), 7.53 (d, J=5.8O Hz, 1 H), 7.55 (s, 1 H), 8.22 (d, J=5.49 Hz, 1 H), 10.18 (s, 1 H).
Example 36
Figure imgf000104_0001
Preparation of Compound 4
To a suspension of 49 mg (0.105 mmol) of (lS,4R,6S,\4S,l8R)-7-cis-l4-tert- butoxycarbonylamino- 18-hydroxy-2, 15-dioxo-3 , 16- diazatricyclo[14.3.0.04'6]nonadec-7-ene-4-carboxylic acid (prepared in Ex. 25, step 4) and 26 mg (0.106 mmol) Of LaCl3 in 1.0 mL of DMF cooled to -78 0C was added 0.53 mL (0.53 mmol) of IM KO1Bu in THF, followed by the addition of 4-chloro-8- fluoroquinoline (19 mg, 0.105 mmol). The mixture was stirred for an hour and warmed to rt. Analytical reversed phase HPLC (Method G) showed no starting material but two new products consistent with the displacement at the 4-Cl (MS m/z, [M++l] = 611, retention time 2.78 min, major component), and at the 8-F (MS m/z, [M++l] = 627, retention time 3.20 min, minor component) of the quinoline ring. It was quenched with a half-saturated NH4Cl aq. solution and organic residues extracted into EtOAc (10 mL X 3). The combined EtOAc extracts were dried (MgSO4), concentrated in vacuo and dissolved in 2 mL of MeOH. This solution was separated by preparative HPLC using the following conditions: Column Xterra 30X100 mm S5, 30% to 100% Solvent B/A for 14 min gradient, hold time 5 min; where Solvent A is 10% MeOH/90% H2O with 0.1%TFA, Solvent B is 90% MeOH/10% H2O with 0.1%TFA and flow rate is 40 mL/min). The major component was not recovered from the preparative HPLC while the minor component, ( 15,4/?, 65, 145,1 BR)-I -cis- 14-terf-butoxycarbonylamino-l 8-(4-chloroquinolin-8-yloxy)-2,l 5-dioxo-3, 16- diazatricyclo[14.3.0.04>6]nona-dec-7-ene-4-carboxylic acid, was collected and concentrated into a white foam (1.9 mg, 3%). 1H NMR (400 MHz5 CD3OD) δ 1.02 (s, 9 H), 1.18-1.47 (m, 6 H)5 1.48-1.77 (m, 3 H), 1.93 (m, 1 H), 2.22-2.34 (m, 2 H), 2.44 (m, 1 H)5 2.56-2.64 (m, 1 H), 2.70-2.78 (m, 1 H), 4.02 (m, 1 H), 4.14 (m, 1 H)5 4.54 (m5 1 H)5 5.38 (m, 1 H)5 5.52-5.62 (m, 2 H)5 7.6 (d, 3=9 Hz5 1 H)5 7.86 (t, J= 8 Hz5 1 H), 7.94-8.03 (m, 2 H), 8.9 (d, J=8 Hz5 1 H). LC-MS m/z 627 [M++!]. Analytical LCMS conditions : 3X50mm YMC Xterra, gradient 3 min, flow 4 mL/min, internal record # 1002J.035. Example 37
Figure imgf000105_0001
Figure imgf000105_0002
Step 1: Prepared l-(2(5)-tert-butoxycarbonylamino-non-8-enoyl)-4(i?)- benzyloxy-pyrrolidine-2(-?)-carboxylic acid, methyl ester by way of example 25, step 1 using 5-AUyloxy-2(S)-(tert-butoxycarbonylamino)pentanoic acid (2.77 g, 10.1 mmol; prepared in Ex. 29, step 5) and methyl 4(R)- benzyloxypyrrolidine-2(iS)-carboxylate hydrochloride (2.50 g, 9.22 mmol) to give 1 -(2(iS)-tert-butoxycarbonylamino-non-8-enoyl)-4(i?)-benzyloxy- pyrrolidine-2(iS)-carboxylic acid methyl ester as a colorless oil, 4.53 g (100%), MS 491 (ES+, M+H+).
Figure imgf000106_0001
Step 2: Prepared l-{[l-(2(,S)-te^Butoxycarbonylamino-non-8-enoyl)-4(i?)- benzyloxypyrrolidine-2(S)carbonyl]-(li2)-amino}-2(S)-vinyl- cyclopropanecarboxylic acid ethyl by way of example 25, step 2 using 1- (2(S)-tert-butoxycarbonylarnino-non-8-enoyl)-4(i?)-benzyloxy-pyrrolidine- 2(S)-carboxylic acid methyl ester (2.78g 5.80 mmol), saponifying and then coupling with (li?,25>l-amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride (0.989 g, 6.38 mmol) to give l-{[l-(2(S)-tert- Butoxycarbonylamino-non-8-enoyl)-4(i?)-benzyloxypyrrolidine- 2(ιS)carbonyl]-(l/?)-amino}-2(S)-vinyl-cyclopropanecarboxyIic acid ethyl ester as a light yellow thick oil, 3.21 g (90%), MS 614 (M+ 1).
Figure imgf000106_0002
Step 3: Prepared (1S,4R,6S,14S18R, 7-cj,y)-14-tert-butoxycarbonylamino-18- benzyloxy^lS-dioxo-Sjlό-diazatricyclotM.S.O.O^J-nonadec-V-ene^- carboxylic acid, ethyl ester by way of example 25, step 3 using l-{[l-(2(5)- tert-butoxycarbonylamino-non-8-enoyl)-4(i?)-benzyloxypyrrolidine- 2(iS)carbonyl]-(li?)-amino} -2(5)-vinyl-cyclopropanecarboxylic acid ethyl ester (2.71 g, 4.42 mmol) to give (1S,4R,6$,14S, 18R, l-cis)-\4-tert- butoxycarbonylamino-18-benzyloxy-2,l 5-dioxo-3, 16- diazatricyclo[14.3.0.04>6]-nonadec-7-ene-4-carboxylic acid ethyl ester as a tan foam, 1.44 g (56%), MS 586 (ES+, M+H+).
Figure imgf000107_0001
Step 4: Prepared (1S,4R,6S,14S,18R, 7-cis)-14-tert-butoxycarbonylamino-18- benzyloxy-2,15-dioxo-3,16-diazatricyclo[14.3.0.04'6]-nonadec-7-ene-4- carboxylic acid by way of example 25, step 4 using (IS,4R, 6S, 14S, 1 SR, 7- cis)- 14-tert-butoxycarbonylamino- 18-benzyloxy-2, 15-dioxo-3, 16- diazatricyclo[14.3.0.04'6]-nonadec-7-ene-4-carboxylic acid ethyl ester (1.30 g, 2.22 mmol) to give (\S,4RfiS,\4S,\%R, 7-cis)-14-tert-butoxycarbonylamino- 18-benzyloxy^.15-dioxo-3^ό-diazatricyclofM.3.O.O^J-nonadec^-ene^- carboxylic acid as a white powder, 0.862 g (70%), MS 558 (ES+, M+l).
Figure imgf000107_0002
Step 5: Prepared (1S,4R, 6S, 143, 18R, 7-e/s)-l 8-benzyloxy- 14-tert- butoxycarbonylamino-4-cyclopropanesulfonylaminocarbonyl-2,15-dioxo- 3,16-diaza-10-oxatricyclo[14.3.0.04'6]nonadec-7-ene by way of example 26, step 4 using (IS,4R,6S,14S, 1 BR, 7-cw)-14-?e^-butoxycarbonylamino-18- benzyloxy-2,15-dioxo-3,16-diazatricyclo[14.3.0.04>6]-nonadec-7-ene-4- carboxylic acid (860 mg, 1.51 mmol) and cyclopropylsulfonamide (365 mg, 3.02 mmol) to give (1S,4R,6S,14S, 18R, 7-czs)-l 8-benzyloxy- 14-tert- butoxycarbonylamino-4-cyclopropanesulfonylaminocarbonyl-2,15-dioxo- 3,16-diaza-10-oxatricyclo[14.3.0.04'6]nonadec-7-ene as a white powder, 603 mg (61 %). MS 660 (ES+, M+H+), HRMS cal. 661.2907 found 661.2903, mp 147-149° C, 1H NMR (300 MHz, CDCl3) δ 0.83 - 0.95 (m, 3 H), 1.01 - 1.16 (m, 3 H), 1.21 - 1.27 (m, 2 H), 1.38 (s, 9 H), 1.42 -1.52 (m, 2 H), 1.87-1.93 (m, 1 H), 1.97 - 2.04 (m, 1 H), 2.30-2.36 (m, 1 H), 2.59-2.68 (q, J= 9 Hz 1 H), 2.82-2.91 (m, 1 H), 3.57-3.62 (dd, J= 9 Hz & 3 Hz, 1 H), 3.69-3.74 (dd, J = 9 Hz & 6 Hz, 1 H), 4.19 - 4.23 (d, J= 12 Hz, 1 H), 4.31 - 4.39 (m, 2 H), 4.43 - 4.58 (m, 2 H), 5.18 - 5.25 (t, J= 9 Hz, 2 H), 5.68-5.76 (m, 1 H), 6.73 (s, 1 H), 7.25 - 7.31 (m, 5H), 10.00 (s, 1 H). Example 38
Figure imgf000108_0001
Preparation of Compound 6
Prepared (IS,4R,6S,14$Λ8R, 7-cis)-l$-tert-butoxy-l4-tert- butoxycarbonylamino-4-cyclopropanesulfonylaminocarbonyI-2, 15-dioxo- 3,16-diaza-10-oxatricyclo[14.3.0.04>6]nonadec-7-ene from methyl 4(R)-tert- butoxypyrrolidine-2(-S)-carboxylate hydrochloride employing the procedures of example 37, steps 1-5; MS 627 (ES+, M+H+), HRMS cal. 627.3064, found 627.3073, 1H-NMR (500 MHz, CD3OD) δ 5.77-5.72 (m, IH), 5.43-5.39 (m, IH), 4.58 (br s, IH), 4.54-4.49 (m, 2H), 4.33 (m, IH), 3.94 (m, IH), 3.83 (m, IH), 3.74 (m, IH), 3.57-3.48 (m, 2H), 2.93 (m, IH), 2.63 (m, IH), 2.25 (m, IH), 2.15 (m, IH), 1.99 (m, IH), 1.76 (m, 2H), 1.70-1.67 (m, IH), 1.58 (m, 2H), 1.44 (s, 9H), 1.29 (m, IH), 1.25 (s, 9H), 1.15-1.09 (m, 2H), 1.04 (m, IH). Example 39 through Example 86 describe the preparation of intermediates.
These intermediates can be used to make compounds of formula (I) by using the teachings described, or referenced, in this document. Example 39
Preparation of Intermediate 39;
Figure imgf000109_0001
Scheme 1
Figure imgf000109_0002
Step 1: A mixture of 3,5-dimethyl-4-nitro-isoxazole (1.42 g, 10.0 mmol), phenylacetaldehyde (1.32 g, 11.0 mmol) in piperidine (1 mL) and ethanol (10 mL) was heated to reflux for 16 h. After cooling down to the ambient temperature, the product precipitated out was collected by filtration. The cake was washed with cold ethanol thoroughly to afford 1.20 g (53%) of the desired product as a white solid.1H NMR (CDCl3) δ 2.87 (s, 3H), 7.46-7.50 (m, 3H), 7.56 (d, J=8.5 Hz, IH), 7.7-7.80 (m, 2H); MS m/z 227 (M++H).
Step 2: A solution of 3-methyl-5-phenyl-isoxazolo[4,5-b]pyridine 4-oxide (1.00 g, 4.40 mmol) and POC13 (2.71 g, 17.7 mmol) in chloroform (10 mL) was heated to reflux for 1 h. After cooling down to the ambient temperature, the final solution was diluted with chloroform (50 mL) and washed with NaHCO3 (aq.) (two 50 mL portions) and brine, dried over MgSO4, filtered, evaporated. The residue was purified by flash chromatography (4:1 hexane-EtOAc) to afford 790 mg (73%) of the desired product as a white solid. IH NMR (CDC13) δ 2.72 (s, 3H), 7.46-7.54 (m, 3H), 7.91 (s, IH), 8.00-8.03 (m, 2H); MS m/z 245, 247 (M++H). Intermediate 39 can be used to make compounds of formula (I) as follows:
Figure imgf000110_0001
Example 40
Preparation of Intermediate 40
Figure imgf000110_0002
Figure imgf000110_0003
Step 1: A mixture of 2-amino-6-methylpyridine (1.08 g, 10.0 mmol), ethyl benzoylacetate (2.30 g, 12.0 mmol) and polyphosphoric acid (6.00 g, 61.2 mmol) was heated to 1100C for 5 h. After cooling to the ambient temperature, the mixture was poured into iced water (20 mL) and neutralized to pH 7 with 10 M NaOH. Extracted with CHCI3. The organic layer was washed with brine, dried over MgSO4, filtered, evaporated. The residue was purified by flash chromatography (1 : 1 hexane-EtOAc) to afford 510 mg (22%) of the desired product as a pale yellow solid. 1H NMR (CDCl3) δ 3.08 (s, 3H), 6.64 (d, J=7.0 Hz, IH), 6/71 (s, IH), 7.42-7.52 (m, 5H), 8.04- 8.06 (m, 2H); MS m/z 237 (M++H).
Step 2: A solution of 6-methyl-2-phenyl-pyrido[l,2a]pyrimidin-4-one (489 mg, 2.07 mmol) in melted diphenyl ether (5 mL) was heated to gentle reflux for 5 h. After cooling to the ambient temperature, the formed suspension was diluted with diethyl ether (10 mL), filtered. The cake was washed with diethyl ether thoroughly to afford 450 mg (92%) of the desired product as a brownish solid.MS m/z 237 (M++H). Step 3: A suspension of 7-methyl-2-phenyl-lH-[l,8]naphthyridin-4-one (450 mg, 1.91 mmol) in POCl3 (10 mL) was heated to gentle reflux for 3 h. then evaporated in vacuo. The residue was poured into iced water (20 mL) and neutralized to pH 10 with 10 M NaOH. The mixture was then extracted with CHCl3 and the organic layer was washed with brine, dried over MgSO4, filtered and evaporated. The residue was purified by flash chromatography (2: 1 hexane-EtOAc) to afford 450 mg (92%) of the desired product as a pink solid. 1H NMR (CD3OD) δ 2.80 (s, 3H), 7.54-7.56 (m, 3H)5 7.61 (d, J=8.4 Hz, IH), 8.25-8.30 (m, 3H), 8.58 (d, J=8.4 Hz3 IH); MS m/z 255, 257 (M++H). Intermediate 40 can be used to make of formula (I) as follows:
Figure imgf000111_0001
Example 41
Preparation of Intermediate 41
Figure imgf000111_0002
Scheme 1
Figure imgf000112_0001
Figure imgf000112_0002
Step 1: To a solution of 4-methoxyphenethyl alcohol (1.52 g, 10.0 mmol) in CH2C12 (50 mL) at 0OC was added Dess-Martin reagent (4.45 g, 10.5 mmol) in one portion. The formed mixture was allowed to warm to the ambient temperature for 1 h. Washed with sat. Na2S2O3 (aq) and IM NaOH, brine respectively. Dried over MgSO4, evaporated in vacuo to give 1.50 g (100%) of the desired aldehyde as a viscous oil. This product was used as crude without any further purification. Step 2: A solution of 3,5-dimethyl-4-nitro-isoxazole (142 mg, 1.0 mmol), 4- methoxy-phenylacetaldehyde from Example 3, Step 1 (180 mg, 1.1 mmol) in piperidine (0.1 mL) and ethanol (2 mL) was heated to reflux for 12 h. After cooling down to the ambient temperature, the product precipitated out was collected by filtration. The cake was washed with cold ethanol thoroughly to afford 130 mg (51%) of the desired product as a grayish solid. 1H NMR (CDCl3) δ 2.88 (s, 3H)5 3.87 (s, 3H), 7.02 (d, J=8.5 Hz, 2H), 7.50 (d, J=9.0 Hz, IH), 7.57 (d, J=9.0 Hz, IH), 7.81 (d, J=8.5 Hz, 2H); MS m/z 257 (M++H).
Step 3: This product was prepared by the same procedure as described in Example 39, Step 2. 1H NMR (CDCl3) δ 2.70 (s, 3H), 3.87 (s, 3H), 7.00-7.03 (m, 2H), 7.84 (s, IH), 7.96-7.98 (m, 2H); MS m/z 275, 277 (M++H). Intermediate 41. can be used to make compounds of formula (I) as follows:
Figure imgf000113_0001
Example 42
Preparation of Intermediate 42
Figure imgf000113_0002
Figure imgf000113_0003
Step 1 & 2:
Figure imgf000113_0004
This product was prepared by the same procedure as described in Example 41, Step 1& 2, except using 4-fluorophenethyl alcohol instead. MS m/z 245 (M++H). Step 3:
Figure imgf000114_0001
This product was prepared by the same procedure as described in step 2 of Example 39.1H NMR (CDCl3) δ 2.71 (s, 3H), 7.17-7.20 (m, 2H), 7.86 (s, IH), 8.00- 8.02 (m, 2H); MS m/z 263, 265 (M++H). Intermediate 42 can be used to make compounds of formula (I) as follows:
Figure imgf000114_0002
Example 43
Preparation of Intermediate 43
Figure imgf000114_0003
Figure imgf000115_0001
Step 1& 2:
Figure imgf000115_0002
This product was prepared by the same procedure as described in Example 41, Step 1& 2, except using 3-rnethoxy-phenethyl alcohol as starting material. MS m/z 257 (M++H). Step 3:
Figure imgf000115_0003
This product was prepared by the same procedure as described in Example 39 step2. 1H NMR (CDCl3) δ 2.72 (s, 3H), 3.90 (s, 3H), 7.00-7.02 (m, IH), 7.41 (t,
J=8.0 Hz, IH)5 7.55 (d, J=7.5 Hz, IH), 7.59 (d, J=2.0 Hz, IH), 7.89 (s, IH); MS m/z
275, 277 (M++H).
Intermediate 43 can be used to make compounds of formula (I) as follows:
Figure imgf000116_0001
Example 44
Preparation of Intermediate 44
Figure imgf000116_0002
Figure imgf000116_0003
Step 1 & 2:
Figure imgf000116_0004
This product was prepared by the same procedure as described in Example 41, Step 1&2, except using 2-methoxy-phenethyl alcohol as starting material. MS m/z 257 (M-H-H). Step 3:
Figure imgf000117_0001
This product was prepared by the same procedure as described in Example 39, Step 2. 1H NMR (CDCl3) δ 2.721 (s, 3H), 3.88 (s, 3H), 7.03 (d, J=S.O Hz, IH), 7.11 (t, J=7.5 Hz, IH), 7.41-7.44 (m, IH), 7.79-7.81 (m, IH), 8.04 (s, IH); MS m/z 275, 277 (M++H). Intermediate 44 can be used to make compounds of formula (I) as follows:
Figure imgf000117_0002
Example 45
Preparation of Intermediate 45
Figure imgf000117_0003
Intermediate 45 is commercially available.
Intermediate 45 can be used to make compounds of formula (I) as follows:
Figure imgf000118_0001
Example 46
Preparation of Intermediate 46
Figure imgf000118_0002
Interemediate 46 was prepared as described by P. Ferrarini et al, in J Heterocyclic Chem, 1983, plO53. Intermediate 46 can be used to make compounds of formula (I) as follows:
Figure imgf000118_0003
Example 47 Preparation of Intermediate 47
Figure imgf000118_0004
Intermediate 47 was prepared as described by R. Nesi et al, Synth Comm. 1992, 22(16), 2349. Intermediate 47 can be used to make compounds of formula (I) as follows:
Figure imgf000119_0001
Example 48
Preparation of Intermediate 48.
Figure imgf000119_0002
Scheme 1
Figure imgf000119_0003
Figure imgf000119_0004
Figure imgf000119_0005
Step 1:
To a solution of 2-bromo-5-methoxybenzoic acid (1.68g, 7.27mmol) in DMF (5OmL) in a medium pressure flask (Chemglass) was added benzamidine (1.25g, 8.00mmol), K2CO3 (6.Og5 43.6mmol), and copper powder (336mg, 1.45mmol). The reaction mixture was heated to 180C° for Ih. Copper and excess K2CO3 were removed by vacuum filtration and washed with MeOH. The filtrate was concentrated and the resulting crude was purified by flash column chromatography (SiO2, 5% MeOH in DCM) to give a light green solid (1.55g, 84% yield): 1H NMR (DMSO-d6) δ 3.84 (s, 3H), 7.26 (d, J- 7.8 Hz3 IH), 7.46 (br s, 5H), 7.57 (s, IH), 8.38 (br s, 1 H); MS m/z (MH+) 253.
Step 2: To a 0 0C slurry of Boc-c/s-Hydroxyproline-OMe (2.Og, 8.15mmol)and 3 (2.26g, 8.97mmol) in THF (82mL) was added Ph3P and diisopropyl azocarboxylate (1.98 g, 8.97mmol). After stirring at rt for 17h, the reaction mixture was diluted with EtOAc (10OmL) and washed with H2O (5OmL). The aqueous layer was separated and back-extracted with EtOAc (2 x 5OmL). The combined organic layer was washed with brine, dried over MgSO4 and concentrated to give a viscous oil which was redissolved in minimal amount of EtOAc and hexanes was added to effect the precipitation of most of the Ph3PO by-product. Ph3PO was removed by vacuum filtration and the liquid filtrate was concentrated. The resulting viscous oil was purified by a flash column chromatography (Siθ2, 4: 1 hex:EtOAc) to give a white solid product (1.76g, 45% yield): 1H NMR (60/40 rotomers, CDCl3) 6 1.47 (s, 9H), 2.49-2.55 (m, IH), 2.73-2.83 (m, IH), 3.80 (s, 1.8H), 3.81 (s, 1.2H)5 3.96 (s, 3H), 4.03-4.09 (m, IH)5 4.54 (t, J- 8.0 Hz, 0.6H), 4.66 (t, J= 7.8 Hz), 4.96-5.06 (m, IH)5 5.97 (br s, 0.6H), 6.04 (br s, 0.4H), 7.33 (dd5 J= 6.1, 2.7 Hz5 IH), 7.46-7.51 (m, 4H)5 7.91 (d, J= 9.2 Hz5 IH), 8.49 (t, J= 8.5Hz5 2H); 13C NMR (rotomers, CDCl3) δ
21.7, 22.O5 28.3, 28.4, 35.8, 36.8, 52.3, 52.4, 52.6, 55.8, 55.9, 57.9, 58.3, 74.5, 74.9, 80.6, 101.2, 101.3, 115.7, 125.8, 126.0, 128.1, 128.5, 129.7, 130.2, 137.9, 147.8, 153.8, 157.7, 158.0, 158.0, 164.8, 173.1, 173.3; MS m/z (MH+) 480. Intermediate 48 can be used to make compounds of formula (I) as follows:
Figure imgf000121_0001
Example 49
Preparation of Intermediate 49.
Figure imgf000121_0002
Stepl:
As described for Example 48
Figure imgf000121_0003
Data: 1H NMR (DMSO-d6) δ 0.97-1.01 (m, 2H), 1.03-1.06 (m, 2H), 1.90-1.94 (m, IH), 3.84 (s, 3H), 3.87 (s, 3H), 6.93 (s, IH), 7.37 (s, 3H), 12.28 (s, IH); 13C NMR (DMSOd6) 59.03, 13.17, 55.47, 55.73, 104.81, 107.27, 113.26, 145.16, 147.48, 154.44, 157.21, 160.89 ; MS m/z (MH+) 247. Step 2:
As described for Example 48.
Figure imgf000122_0001
Data: 1H NMR (CDCl3) δ 1.00-1.04 (m, 2H)5 1.07-1.11 (m, 2H), 1.43 (s, 5.4H), 1.46 (s, 3.6H), 2.17-2.21 (m, IH), 2.37-2.43 (m, IH), 2.62-2.69 (m, IH), 3.75 (s, 1.8H), 3.78 (s, 1.2H), 3.92 (d, J- 2.8 Hz, IH), 4.00 (s, 3.6H), 4.01 (s, 2.4H), 4.48 (t, J- 8.0 Hz, 0.6H), 4.59 (t, J= 7.6 Hz, 0.4H), 5.7 (br s, 0.6H), 5.74 (br s, 0.4H), 7.18 (s, IH), 7.20 (s, IH); 13C NMR (CDCl3) δ 9.6, 9.7, 18.1, 28.3, 28.4, 35.8, 36.7, 52.2, 52.4, 56.3, 57.8, 58.2, 74.0, 74.5, 80.5, 80.6, 101.0, 101.1, 106.3, 108.6, 148.8, 149.1, 153.8, 155.4, 164.4, 165.9, 172.9, 173.2; LC-MS m/z (MH+) 474. Intermediate 49 can be used to make compounds of formula (I) as follows:
Figure imgf000122_0002
Example 50
Preparation of Intermediate 50.
Figure imgf000122_0003
Step 1:
As described in Example 48 wherein acetamidine hydrochloride and 2-bromo-5- methoxybenzoic acid were utilized as starting materials.
Product:
Figure imgf000123_0001
Data: 1H NMR (DMSO) δ 2.31 (s, 3H), 3.85 (s, 3H), 7.36 (d, J = 6.2 Hz, IH), 7.37 (s, IH), 7.51 (d, J = 7.8 Hz, IH), 12.15 (s, IH); 13C NMR (DMSO) 521.11, 55.41, 105.57, 121.22, 123.59, 128.12, 143.34, 151.68, 157.00, 161.45; LC-MS m/e (MH+) 191. Step 2: As described in Example 48.
Figure imgf000123_0002
Data: 1H NMR (CDCl3) δ 1.43 (s, 5.4H), 1.45 (s, 3.6H), 2.38-2.45 (m, IH), 2.62- 2.71 (m, IH), 2.66 (s, 1.8H), 2.68 (s, 1.2H), 3.77 (1.8H), 3.79 (s, 1.2H), 3.92 (s, 3H), 3.93-3.98 (m, 2H), 4.49 (t, J = 8.0 Hz, 0.6H), 4.61 (t, J = 7.8 Hz, 0.4H), 5.82 (t, J = 2.1 Hz, 0.6H), 5.89 (t, J = 2.3 Hz, 0.4H), 7.26 (dd, J = 4.7, 3.2 Hz, IH), 7.42 (dd, J = 6.3, 2.8 Hz, IH), 7.75 (d, J = 9.15 Hz, IH); 13C NMR (CDCl3) δ 26.1, 28.3, 28.4, 35.8, 36.7, 52.2, 52.2, 52.4, 52.5, 55.755.8, 57.9, 58.2, 74.1, 74.7, 80.6, 101.0, 101.2, 114.9, 125.6, 125.9, 128.6, 147.3, 153.8, 154.5, 157.6, 157.6, 161.2, 164.6, 173.0, 173.3; LC- MS m/e (MH+) 418. Intermediate 50 can be used to make compounds of formula (I) as follows:
Figure imgf000124_0001
Example 51
Preparation of Intermediate 51.
Figure imgf000124_0002
Stepl: Prepared as described in Example 48 and using 2-bromo-4,5- dimethoxybenzoic acid and trifluoroamidine as starting materials.
Figure imgf000124_0003
Data: 1H NMR (DMSO) 5 3.92 (s, 3H)1, 3.94 (s, 3H), 7.33 (s, IH), 7.50 (s, IH), 13.40 (br s, IH); 13C NMR (DMSO) δ 55.8, 56.1, 104.9, 108.7, 150.2, 155.0; LC- MS m/e (MH+) 275.
Step 2: As described in Example 48. Product:
Figure imgf000124_0004
Data: 1H NMR (CDCl3) δ 1.42 (s, 3.6H), 1.44 (s, 5.4H), 2.42-2.49 (m, IH), 2.67- 2.73 (m, IH), 3.37 (s, 1.2H), 3.78 (s, 1.8H), 3.97 (t, J = 6.5 Hz, IH), 4.02 (s, 2.4H), 4.04 (s, 3.6H), 4.48 (t, J = 7.9 Hz, 0.6H), 4.60 (t, J = 7.7 Hz, 0.4H), 5.86 (br s, 0.6H), 5.90 (br s, 0.4H), 7.27-7.29 (m, IH), 7.38-7.44 (m, IH); 13C NMR (CDCl3) δ 8.2, 28.3, 35.7, 36.7, 52.1, 52.2, 52.4, 56.5, 57.8, 58.2, 75.5, 76.0, 80.7, 100.8, 107.6, 111.0, 119.7, 148.2, 150.2, 151.4, 153.8, 154.5, 156.4, 165.1, 172.7, 173.0; LC-MS m/e" (MH+) 502.
Intermediate 51 can be used to make compounds of formula (I) as follows:
Figure imgf000125_0001
Example 52
Preparation of Intermediate 52.
Figure imgf000125_0002
Intermediate 52 is commercially available and can be used to make compounds of formula (I).
Figure imgf000125_0003
Example 53
Preparation of Intermediate 53
Figure imgf000126_0001
Intermediate 53 is commercially available and can be used to make compounds of formula (I).
Figure imgf000126_0002
Compound of Formula I
Example 54
Preparation of Intermediate 54
Figure imgf000126_0003
Intermediate 54 is commercially available and can be used to make compounds of formula (I).
Figure imgf000126_0004
Example 55
Preparation of Intermediate 55
Figure imgf000127_0001
Reference scheme for preparation of Intermediate 55. Scheme 1
Figure imgf000127_0002
Step 1 : A solution of 3-phenyl-but-2-enoic acid (16.2 g), diphenylphosphoryl azide (27.5 g), and triethylamine (10.1 g) in benzene (100 mL) was stirred for 1 h. After filtration through a silica gel plug washing with benzene and concentration, the residue was dissolved in diphenylmethane (80 mL) and refiuxed for 3 h. After cooling to rt, solids were collected through a plug washing with benzene and dried to give 10 g (63%) of the desired product as a solid. 1H NMR (400 MHz, CD3OD) δ ppm 2.30 (s, 3 H), 7.00 (s, 1 H), 7.54 (m, 1 H), 7.77 (m, 2 H)5 8.33 (d, J=7.34 Hz, 1 H). Step 2: A solution of 4-methyl-2H-isoquinolin-l-one (4.8 g) in POCl3 (50 mL) was refiuxed for 3 h. After cooling and concentration, the residue was based with 5 N NaOH and extracted with CH2Cl2. The organic layer was washed with brine and dried over MgSO4. After concentration, purification by flash chromatography of Biotage with 5% ethyl acetate in hexanes gave 4.8 g (90%) of the desired product as a solid. 1H NMR (400 MHz, CDCl3) δ ppm 2.59 (s, 3 H), 7.68 (t, J=7.70 Hz, 1 H), 7.78 (m, 1 H), 7.94 (d, J=8.31 Hz, 1 H), 8.11 (s, 1 H), 8.35 (d, J=8.31 Hz, 1 H). Chemistry for preparation of Intermediate 55.
Figure imgf000127_0003
Step 1: Preparation of 7-fluoro-6-methoxy-2/ϊ-isoquinolin-l-one. As shown in step 1 of this example using 19.6 g 4-fluoro-3-methoxycinnamic acid as starting material. 9.5 g product obtained (48% yield).
Figure imgf000128_0001
Data: 1H NMR (400 MHz, CD3COCD3) δ ppm 4.00 (s, 1 H), 6.49 (d, J=7.34 Hz, 1 H), 7.19 (d, J=7.09 Hz, 1 H), 7.29 (d, J=8.07 Hz, 1 H), 7.86 (d, J=I 1.74 Hz, 1 H). Step 2: Preparation of l-chloro-7-fluoro-6-methoxyisoquinoline: As shown in step 2 of this example using 7-fluoro-6-methoxy-2H-isoquinolin-l-one (9g) as starting material. 7 g of desired product obtained (70% yield).
Figure imgf000128_0002
Data: 1H NMR (400 MHz, CDCl3) δ ppm 4.04 (s, 3 H), 7.17 (d, J=8.07 Hz, 1 H), 7.48 (d, J=5.62 Hz, 1 H), 7.94 (d, J=I 1.49 Hz, 1 H), 8.20 (d, J=5.62 Hz, 1 H). Intermediate 55 can be used to make compounds of formula (I).
Figure imgf000128_0003
Example 56
Preparation of Intermediate 56.
Figure imgf000128_0004
Step 1: As in Example 55 step 1 but with 3.82 g of 3-(4-Fluoro-phenyl)-3-methoxy- acrylic acid as starting material. 198 mg product obtained (5% yield). Product:
Figure imgf000129_0001
Data: MS: (M+H)+ 194.
Step 2: As in Example 55, step 1 , but with 193 mg 7-fluoro-4-methoxy-2H- isoquinolin-1-one as starting material. 199 mg product obtained (94% yield).
Product:
Figure imgf000129_0002
Data: 1H NMR (400 MHz, CDCl3) δ ppm 4.05 (s, 3 H), 7.49 (m, 1 H), 7.78 (s, 1 H), 7.86 (dd, J=9.66, 2.57 Hz, 1 H), 8.23 (dd, J=9.29, 5.38 Hz, 1 H); MS: (M+H)+ 212. Intermediate 56 can be used to make compounds of formula (I).
Figure imgf000129_0003
Example 57
Preparation of Intermediate 57.
Figure imgf000129_0004
Intermediate 57 can be used to make compounds of formula (I).
Figure imgf000130_0001
Example 58
Preparation of Intermediate 58.
Figure imgf000130_0002
Figure imgf000130_0003
To a solution of B oc-cis-H YP-OMe (122.6 mg, 0.5 mmol) in THF (15 mL) at 00C, triphenylphosphine (196.7 mg, 0.75 mmol) and benzo[d]isoxazol-3-ol (81 mg, 0.6 mmol) were added. Then DEAD (0.118 mL, 0.75 mmol) was added. The reaction mixture was warmed to rt. and stirred for 3 hr. Then solvent was evaporated and the residue was purified by Prep. HPLC to give a colorless thick oil. (117 mg, 54% yield) 1H NMR (400 MHz, CD3OD) δ 1.41 (m, 9 H), 2.38 (m, 1 H), 2.75 (m, 1 H), 3.75 (m, 3 H)5 3.81 (m, 1 H), 3.90 (m, 1 H), 4.47 (m, 1 H), 5.44 (m, 1 H), 7.31 (t, J=7.46 Hz, 1 H), 7.47 (d, J=8.56 Hz, 1 H), 7.59 (t, J=7.83 Hz, 1 H)5 7.66 (d, J=8.07 Hz, 1 H). LC-MS (retention time: 2.65 min.), MS m/z 363(MH+). Intermediate 58 can be used to make compounds of formula (I).
Figure imgf000131_0001
I
Example 59
Preparation of Intermediate 59.
Figure imgf000131_0002
To a solution of 2,4-dichloropyrimidine (149 mg, 1 mmol) in THF (5 mL), tetrakis(triphenylphosphine) palladium (23 mg, 2 mol%) and 0.5M solution of phenylzinc bromide (2.1 mL. 1.05 mmol) in THF were added. The reaction mixture was stirred at 500C for overnight. Then it was added saturated ammonium chloride solution and extracted with EtOAc twice. The organic layers were combined, washed with water and dried (MgSO4). Evaporation of solvent gave a yellow residue which was purified by Prep. HPLC to afford a yellowish oil as 2-chloro-4-phenyl- pyrimidine. Intermediate 59 can be used to make compounds of formula (I).
Figure imgf000131_0003
Example 60
Preparation of Intermediate 60.
Figure imgf000132_0001
To a solution of 2,4-dichloropyrimidine (149 mg, 1 mmol) in THF (5 mL), tetrakis(triphenylphosphine) palladium (58 mg, 5 mol%) and 0.5M solution of 2- pyridinylzinc bromide (2.4 mL, 1.2 mmol) in THF were added. The reaction mixture was stirred at 500C for overnight. Then it was added saturated ammonium chloride solution and extracted with EtOAc twice. The organic layers were combined, washed with water and dried (MgSO4). Evaporation of solvent gave a yellow residue which was purified by Prep. HPLC to afford a yellowish oil as product. (Intermediate 60, 11 mg, 3.6 % yield) 1H NMR (500 MHz, CD3OD) δ 7.61 (m, 1 H), 8.07 (m, 1 H), 8.36 (d, J=5.19 Hz, 1 H), 8.50 (d, J=7.94 Hz, 1 H), 8.75 (d, J=3.97 Hz, 1 H)5 8.82 (d, J=5.19 Hz, 1 H). MS m/z 192 (MH+). Intermediate 60 can be used to make compounds of formula (I).
Figure imgf000132_0002
Example 61
Preparation of Intermediate 61.
Figure imgf000133_0001
To a solution of 2,4-dichloropyrimidine (149 mg, 1 mmol) in DMF (5 mL), dichloro bis(triphenylphosphine) palladium (II) (35 mg, 5 mol%) and 2- (tributylstannyl)thiophene (0.38 mL, 1.2 mmol) were added. The reaction mixture was heated at 7O0C for 3hr. Then it was added saturated KF solution in methanol (20 mL) and stirred at rt for 4 hr. The reaction mixture was concentrated with a small amount of silica gel and the residue was filtered through filter paper and washed with EtOAc. The filtrate was then concentrated and the residue was purified by Prep. HPLC to afford an off-white solid as product. (110 mg, 35 % yield) 1H NMR (400 MHz, CD3OD) δ 7.20 (dd, J=5.01, 3.79 Hz, 1 H), 7.74 (dd, J=5.01, 1.10 Hz, 1 H), 7.77 (d, J=5.38 Hz, 1 H), 7.98 (dd, J=3.79, 1.10 Hz, 1 H)5 8.55 (d, J=5.38 Hz, 1 H). MS m/z 197 (MH+). Intermediate 61 can be used to make compounds of formula (I).
Figure imgf000133_0002
Example 62
Preparation of Intermediate 62.
Figure imgf000134_0001
To a solution of 2,4-dichloropyrimidine (149 mg, 1 mmol) in DMF (5 mL), dichloro bis(triphenylphosphine) palladium (II) (35 mg, 5 mol%) and 2-
(tributylstannyl)furan (0.35 mL, 1.1 mmol) were added. The reaction mixture was heated at 700C for 3hr. Then it was added saturated KF solution in methanol (20 mL) and stirred at rt for 4hr. The reaction mixture was concentrated with a small amount of silica gel and the residue was filtered through filter paper and washed with EtOAc. The filtrate was then concentrated and the residue was purified by Prep. HPLC to afford a brownish solid as product. (80 mg, 27 % yield) 1H NMR (400 MHz, CD3OD) δ 6.68 (dd, J=3.67, 1.71 Hz, 1 H), 7.42 (d, J=3.67 Hz, 1 H), 7.67 (d, J=5.13 Hz, 1 H), 7.30 (d, .7=1.71 Hz, 1 H),~8.62 (d, J=5.14 Hz, 1 H). MS m/z 181 (MH+). Intermediate 62 can be used to make compounds of formula (I).
Figure imgf000134_0002
Example 63
Preparation of Intermediate 63.
Figure imgf000135_0001
To a solution of 2,4-dichloropyrimidine (149 mg, 1 rnmol) in DMF (5 mL), dichloro bis(triphenylphosphine) palladium (II) (35 mg, 5 mol%) and 2-
(tributylstannyl)thiazole (412 mg, 1.1 mmol) were added. The reaction mixture was heated at 800C for 3hr. Then it was added saturated KF solution in methanol (20 mL) and stirred at rt for 4hr. The reaction mixture was concentrated with a small amount of silica gel and the residue was filtered through filter paper and washed with EtOAc. The filtrate was then concentrated and the residue was purified by Prep. HPLC to afford a brownish solid as product. (9 mg, 3 % yield). MS m/z 198 (MH+). Intermediate 63 can be used to make compounds of formula (I).
I
Figure imgf000135_0002
Example 64 Preparation of Intermediate 64.
Figure imgf000135_0003
Scheme 1
Figure imgf000136_0001
Step 1 : To a solution of Boc-HYP-OH (1.0 g, 4.324 mmol) in DMF (20 mL), NaH (0.38 g of 60% dispersion in mineral oil, 9.513 mmol) was added at 00C. The reaction mixture was stirred for 1 hr. Then 2,4-dichloropyrimidine (0.709 g, 0.0289 mmol) was added. The reaction mixture was warmed to rt and stirred for overnight. It was then quenched with IN HCl solution and extracted with EtOAc. The organic layer was separated, washed with brine and dried (MgSO4). Evaporation of solvent gave crude product which was then purified by Prep. HPLC to give colorless oil as product. (0.4 g, 27% yield) 1H NMR(CD3OD, 300 MHz) 6 1.13 (m, 9 H), 2.37 (m, 1 H), 2.62 (m, 1 H)5 3.70-3.84 (m, 2 H), 4.38 (m, 1 H), 5.65 (m, 1 H), 6.88 (d, J=5.86 Hz, 1 H), 8.37V5 J=5-86 Hz5 1 H). MS m/z 344(MH+).
Step 2: To a solution of (2S, 4R) 4-(2-Chloro-pyrimidin-4-yloxy)-pyrrolidine-l,2- dicarboxylic acid 1-tert-butyl ester (0.34 g, 0.99 mmol) in CH3CN (20 mL) was added (IR, 2S)/(1S, 2R) (l-cyclopropanesulfonyl-aminocarbonyl-2-vinyl-cyclo- propyl)-carbamic acid (0.511 g, 1.48 mmol), DIEA (0.86 mL, 4.95 mmol) and the coupling reagent HOBt (0.226 g, 1.48 mmol) and HBTU (0.561 g, 1.48 mmol). The solution was stirred at rt. overnight. Then it was concentrated, washed with water and extracted with ethyl acetate twice. The combined organic layers were washed with brine, dried over MgSO4 and concentrated. It was then purified by Prep. HPLC column to give a yellow solid (A). (0.33 g, 41% yield). MS m/z 655 (MH+). Step 3: To a solution of intermediate 4 (50 mg, 0.061 mmol) in CH2Cl2 (2.5 mL), 1,2,3,4-tetrahydroisoquinoline (0.011 mL, 0.0915 mmol) and Et3N (0.021 mL, 0.153 mmol) were added. The reaction mixture was stirred at rt for overnight and at 4O0C for 1 day. The solvent was stripped and the residue was purified by Prep. HPLC to give a colorless oil. It was then dissolved in 4N HCl in dioxane (1 mL) and stirred for overnight. Evaporation of solvent gave a colorless oil as hydrochloride salt. (20 mg, 52% yield). MS m/z 553 (MH+). Step 4: To a solution of 4-[2-(3,4-Dihydro-lH-isoquinolin-2-yl)-pyrimidin-4-yloxy]- pyrrolidine-2-carboxylic acid (l-cyclopropanesulfonylaminocarbonyl-2-vinyl- cyclopropyl)-amide hydrochloride (20 mg, 0.032 mmol) in CH3CN (5 mL) was added 2-methoxycarbonylamino-3,3-dimethyl-butyric acid (9.1 mg, 0.048 mmol), DIEA (0.028 mL, 0.16 mmol) and the coupling reagent HOBt (7.3 mg, 0.048 mmol) and HBTU (18.2 mg, 0.048 mmol). The solution was stirred at rt. overnight. Then it was concentrated, washed with water and extracted with ethyl acetate twice. The combined organic layers were washed with brine, dried over MgSO4 and concentrated to give yellowish oil. It was purified by Prep. HPLC column to give a colorless oil as TFA salt (Intermediate 64). (16 mg, 60% yield) 1H NMR(CD3OD, 500 MHz) § 0.98-1.06 (m, 13 H), 1.13 (m, 1 H), 1.22-1.32 (m, 1 H), 1.35-1.44 (m, 1 H), 1.82 (dd, 7=8.24, 5.19 Hz, 0.5 H), 1.90 (dd, j=8.24, 5.49 Hz, 0.5 H), 2.26 (m, 1 H), 2.32-2.43 (m, 1 H), 2.56 (m, 1 H)5 2.96 (m, 1 H), 3.11 (m, br, 2 H)5 3.56 (s5 3 H)5 4.14 (m5 1 H), 4.21 (m, 1 H)5 4.38 (m, 1 H), 4.47 (m, 1 H), 5.15 (m, 1 H), 5.31 (m, 1 H), 5.75 (m, 1 H), 5.94 (s, 1 H), 6.47 (d, J=7.02 Hz, 1 H), 7.29 (s, 4 H), 7.49 (m, 1 H), 7.56 (m, 1 H)5 7.74 (d, J=8.24 Hz, 1 H)5 7.88 (d, J=8.24 Hz5 1 H)5 8.11 (d, J=7.02
Hz, l.H).MS m/z 724 (MH+).
Intermediate 64 can be used to make compounds of formula (I).
Figure imgf000138_0001
Example 65
Preparation of Intermediate 65.
Figure imgf000138_0002
To a solution of A (50 mg, 0.061 mmol) in CH2Cl2 (2.5 mL), isoindoline (0.013 mL, 0.115 mmol) and Et3N (0.026 mL, 0.19 mmol) were added. The reaction mixture was stirred at rt for 2 days. The solvent was stripped and the residue was purified by Prep. HPLC to give a colorless oil. It was then dissolved in 4N HCl in dioxane (1 mL) and stirred for overnight. Evaporation of solvent gave crude product which was purified by Prep.HPLC again to afford yellowish solid as TFA salt. (8.5 mg, 14% yield). MS m/z 539 (MH+).
Intermediate 65 can be used to make compounds of formula (I).
Figure imgf000139_0001
Example 66
Preparation of Intermediate 66.
Figure imgf000139_0002
. To a solution of A of Example 64 (50 mg, 0.061 mmol) in CH2Cl2 (2.5 mL), morpholine (0.008 mL, 0.0915 mmol) and Et3N (0.021 mL, 0.153 mmol) were added. The reaction mixture was stirred at rt for overnight and at 400C for 1 day. The solvent was stripped and the residue was purified by Prep. HPLC to give a colorless oil. It was then dissolved in 4N HCl in dioxane (1 mL) and stirred for overnight. Evaporation of solvent gave a colorless oil as hydrochloride salt. (12.6 mg. 36% yield); MS m/z 507 (MH+). Intermediate 66 can be used to make compounds of formula (I).
Figure imgf000139_0003
Example 67 Intermediate 61
Figure imgf000140_0001
Preparation of Intermediate 67
Figure imgf000140_0002
To a solution of 1, 4-p-tolylsulfanylcarbonyl-pyrrolidine-l,2-dicarboxylic acid 1-tert-butyl ester 2-methyl ester (3.0 g, 7.91 mmol)in ethanol (15 mL) and THF (30 mL) mixture, sodium borohydride (0.6 g, 15.8 mmol) was added. The reaction mixture was stirred at rt. for overnight. Then it was concentrated, washed with 1 N HCl solution and extracted with EtOAc three times. The organic layers were combined, washed with saturated NaHCO3 solution and dried (MgSO4). Evaporation of solvent gave yellowish oil which was purified by flash column chromatography (silica gel, 3:1 EtOAc: Hexanes) to afford colorless oil as product (2). (1.77 g, 86% yield) 1H NMR (CD3OD5 500 MHz) $ 1.43 (m, 9 H), 2.00-2.13 (m, 2 H), 2.46 (m, 1 H), 3.19 (m, 1 H), 3.47-3.53 (m, 2 H)5 3.61 (m, 1 H)5 3.73 (m, 3 H), 4.31 (m, 1 H).MS m/z 282 (M+Na+).
To a solution of 2 (80 mg, 0.309 mmol) in THF (10 mL) at 00C, triphenylphosphine (121.4 mg, 0.463 mmol) and 4-hydroxyquinoline (67.2 mg, 0.463 mmol) were added. Then DEAD (80.6 mg, 0.463 mmol) was added. The reaction mixture was warmed to rt. and stirred for 2 days. Then solvent was evaporated and the residue was purified by Prep. HPLC to give colorless oil. It was then dissolved in 4N HCl in dioxane (3 mL) and stirred for 2 hr. Evaporation of solvent gave thick colorless oil as bis HCl salt. (110 mg, 99% yield) 1H NMR(500 MHz5 CD3OD) 6 2.52 (m, 1 H). 2.60 (m, 1 H), 3.19 (m, 1 H), 3.45 (m, 1 H)5 3.66 (s, 3 H), 3.86 (m, 1 H)5 4.61-4.75 (m5 3 H)5 7.56 (d, J=6.7 Hz5 1 H), 7.94 (t, J=7.3 Hz, 1 H), 8.10-8.20 (m, 2 H)5 8.55 (d, J=8.2 Hz5 1 H)5 9.07 (d, J=6.7 Hz5 1 H). MS m/z 287 (MH+). Intermediate 67 can be used to make compounds of formula (I).
Figure imgf000141_0001
Example 68
Preparation of Intermediate 68
Figure imgf000141_0003
Figure imgf000141_0002
To a solution of 2 from Example 67 (150 mg, 0.578 mmol) in THF (15 rnL) at O0C5 triphenylphosphine (228 mg, 0.868 mmol) and 3-bromophenol (150 mg, 0.868 mmol) were added. Then DEAD (0.14 mL, 0.868 mmol) was added. The reaction mixture was warmed to rt. and stirred for 2 days. Then solvent was evaporated and the residue was purified by Prep. HPLC to give colorless oil as product. (105 mg, 44% yield). MS m/z 436 (M+Na+). Intermediate 68 can be used to make compounds of formula (I).
Figure imgf000142_0001
Example 69
Preparation of Intermediate 69
Figure imgf000142_0002
To a solution of 4-hydroxymethyl-pyrrolidine-l,2-dicarboxylic acid 1-tert- butyl ester 2-methyl ester (2 of Example 67,300 mg, 1.157 mmol) in THF (15 mL) at 00C, triphenylphosphine (455 mg, 1.735 mmol) and 5-bromo-pyridin-3-ol (prepared according to F.E.Ziegler et al., J.Am.Chem.Soc, (1973), 95, 7458) (302 mg, 1.735 mmol) were added. Then DEAD (0.273 mL, 1.735 mmol) was added. The reaction mixture was warmed to rt. and stirred for 2 days. Then solvent was evaporated and the residue was purified by Prep. HPLC to give a yellowish oil. Then it was dissolved in 4N HCl solution in dioxane (3.0 mL) and stirred for 4 hr. Evaporation of solvent gave crude product which was further purified by Prep. HPLC to afford a yellowish oil as TFA salt. (70 mg, 11% yield) MS m/z 315 (MH+). Intermediate 69 can be used to make compounds of formula (I).
Figure imgf000143_0001
Example 70
Preparation of Intermediate 70
Figure imgf000143_0002
Step 1 : To a solution of 2 from Example 67 (700 mg, 2.7 mmol) in THF (90 mL), methanol (50 mL) and water (12 mL) mixture, lithium hydroxide monohydrate (1700 mg, 2.0 mmol) was added. The reaction mixture was stirred at rt. for overnight. Then it was acidified with IN HCl solution to pH=3 to 5. Extracted with ethyl acetate (2x20 mL) and the organic layers were combined and dried (MgSO4). Evaporation of solvent gave thick colorless oil as product (0.58, 88% yield). 1H NMR (CD3OD, 400 MHz) δ 1. 42 (m, 9 H), 2.00-2.09 (m, 2 H), 2.45 (m, 1 H), 3.17 (m, 1 H), 3.49 (m, 2 H), 3.59 (m, 1 H), 4.24 (m, 1 H). MS m/z 268 (M-HNa+). Step 2: To a solution of the proline carboxylic acid (270 mg, 1.1 mmol) in DMSO (10 mL), potassium t-butoxide (309 mg, 2.75 mmol) was added. The reaction mixture was stirred at rt for lhr. Then 2-Bromo-4-chloro-pyridine (254 mg, 1.32 mmol) was added. The reaction mixture was stirred at rt for overnight. Then it was quenched with water and washed with ethyl acetate. The aqueous layer was separated and acidified with IN HCl solution to pH=3. Extracted with ethyl acetate twice and the organic layers were combined and dried (MgSO4). Evaporation of solvent gave an orange oil. It was then dissolved in methanol and HCl (gas) was bubbled through for 2 min at -78°C. Then the reaction mixture was warmed to rt and stirred for overnight. Evaporation of solvent gave an orange oil as crude to carry on. MS m/z 315 (MH+). Intermediate 70 can be used to make compounds of formula (I).
Figure imgf000144_0001
Example 71
Preparation of Intermediate 71
Figure imgf000144_0002
To a solution of intermediate 3 from Example 70 (270 mg, 1.1 mmol) in DMSO (10 mL), potassium t-butoxide (309 mg, 2.75 mmol) was added. The reaction mixture was stirred at rt for lhr. Then 2,6-dibromopyridine (313 mg, 1.32 mmol) was added. The reaction mixture was. stirred at rt for overnight. Then it was quenched with water and washed with ethyl acetate. The aqueous layer was separated and acidified with IN HCl solution to pH=3. Extracted with ethyl acetate twice and the organic layers were combined and dried (MgSO4). Evaporation of solvent gave an orange oil. It was then dissolved in methanol and HCl (gas) was bubbled through for 2 min at -780C. Then the reaction mixture was warmed to rt and stirred for overnight. Evaporation of solvent gave an orange oil as crude to carry on. MS m/z 315 (MH+). Intermediate 71 can be used to make compounds of formula (I).
Figure imgf000145_0001
Example 72 '
Preparation of Intermediate 72
Figure imgf000145_0002
To a solution of 4-hydroxymethyl-pyrrolidine-l,2-dicarboxylic acid 1-tert- butyl ester 2-methyl ester (2 of Example 61, 300 mg, 1.157 mmol) in THF (15 mL) at 0°C, triphenylphosphine (455 mg, 1.735 mmol) and 5-bromo-pyridin-3-ol (prepared according to F.E.Ziegler et a\., J.Am.Chem.Soc, (1973), 95, 7458) (302 mg, 1.735 mmol) were added. Then DEAD (0.273 mL, 1.735 mmol) was added. The reaction mixture was warmed to rt. and stirred for 2 days. Then solvent was evaporated and the residue was purified by Prep. HPLC to give a yellowish oil. Then it was dissolved in 4N HCl solution in dioxane (3.0 mL) and stirred for 4 hr. Evaporation of solvent gave crude product which was further purified by Prep. HPLC to afford a yellowish oil as TFA salt. (70 mg, 11 % yield). MS m/z 315 (MH+). Intermediate 72 can be used to make compounds of formula (I).
Figure imgf000146_0001
In Examples 68-72, the intermediates described (68-72) and the proposed compounds of formula (I)5 each contain a halopyridine functionality. This functionality can be employed in coupling reactions wherein the halo group is replace with a ring system or alternate functionality. This reaction is well recognized in the art and the following reactions serve as examples of said coupling process. Example 73 Coupling Reaction: Example A
Figure imgf000146_0002
To a solution of A (16 mg, 0.0339 mmol) in DMF (1 mL), 3- thiopheneboronic acid (5.6 mg, 0.044 mmol), tetrakis(triphenylphosphine) palladium (2.0 mg, 0.0017 mmol) and 2M Na2CO3 solution (0.051 mL, 0.1017 mmol) were added. The reaction mixture was heated at 1 lOoC for 4hr. Then it was filtered and washed with methanol. The filtrate was concentrated and purified by Prep.HPLC to give brownish oil as product. (6mg, 37% yield) 1H NMR (CD3OD, 400 MHz) 5 1.05 (s, 9 H), 2.21-2.30 (m, 2 H), 2.95 (m, 1 H), 3.42 (s, 3 H), 3.93 (m, 1 H), 4.01 (m, 1 H), 4.20-4.30 (m, 3 H), 4.60 (dd, J-8.56, 5.87 Hz, 1 H), 7.64 (m, 2 H), 8.12 (m, 1 H) 8.37 (m, 1 H), 8.45 (m, 1 H), 8.75 (s, 1 H). MS m/z 476 (MH+). Example 74
Coupling Reaction: Example B
Figure imgf000147_0001
To a solution of A (20 mg, 0.0423 mmol) in DMF (1 mL), 3- thiopheneboronic acid (7.0 mg, 0.055 mmol), tetrakis (triphenylphosphine) palladium (2.4 mg, 0.00212 mmol) and 2M Na2CO3 solution (0.063 mL, 0.127 mmol) were added. The reaction mixture was heated at 11O0C for 30 hr. Then it was filtered and washed with methanol. The filtrate was concentrated and purified by Prep.HPLC to give brownish oil as product. (10.5 mg, 42% yield) MS m/z 476 (MH+). Example 75 Coupling Reaction: Example C
Figure imgf000147_0002
To a solution of A (20 mg, 0.0423 mmol) in DMF (2 mL), 2- thiopheneboronic acid (7.0 mg, 0.055 mmol), tetrakis(triphenylphosphine) palladium (2.4 mg, 0.00212 mmol) and barium hydroxide (40 mg, 0.127 mmol) were added. The reaction mixture was heated at 1500C in Smith microwave reactor for 110 min. Then it was filtered and washed with methanol. The filtrate was concentrated and purified by Prep. HPLC to give yellowish oil as product. (5.0 mg, 20% yield). MS m/z
476 (MH+).
Example 76
Coupling Reaction: Example D
Figure imgf000148_0001
To a solution of A (20 mg, 0.0423 mmol) in DMF (2 mL), 3-furanboronic acid (6.2 mg, 0.055 mmol), tetrakis(triphenylphosphine) palladium (2.4 mg, 0.00212 mmol) and barium hydroxide (40 mg, 0.127 mmol) were added. The reaction mixture was heated at 15O0C in Smith microwave reactor for 30 min. Then it was filtered and washed with methanol. The filtrate was concentrated and purified by Prep.HPLC to give yellowish oil as product. (12 mg, 49% yield) MS m/z 460 (MH+). Example 77 Coupling Reaction: Example E
Figure imgf000148_0002
To a solution of A (25 mg, 0.053 mmol) in DMF (1 mL), 3-thiopheneboronic acid (8.8 mg, 0.0688 mmol), tetrakis(triphenylphosphine) palladium (3.1 mg, 0.00265 mmol) and 2M Na2CO3 solution (0.080 mL, 0.159 mmol) were added. The reaction mixture was heated at 11O0C for overnight. Then it was filtered and washed with methanol. The filtrate was concentrated and purified by Prep.HPLC to give brownish oil as product. (15 mg, 48% yield) 1H NMR (CD3OD, 500 MHz) δ 1.06 (s, 9 H), 2.20-2.31 (m, 2 H), 2.94 (m, 1 H), 3.55 (s, 3 H), 3.91 (m, 1 H), 3.98 (m,l H), 4.34 (s, 1 H), 4.37-4.46 (m, 2 H), 4.61 (dd, J=8.85, 5.19 Hz, 1 H), 6.77 (d, J=8.24 Hz. 1 H), 7.39 (d, J=7.32 Hz, 1 H), 7.48 (dd, J=5.19, 3.05 Hz, 1 H), 7.68 (dd, J=4.88, 1.22 Hz, 1 H), 7.77(t, J=7.93 Hz, IH), 8.04 (m, 1 H). MS m/z 476 (MH4). Example 78 Coupling Reaction: Example F
Figure imgf000149_0001
To a solution of A (20 mg, 0.0423 mmol) in DMF (1 mL), phenyl boronic acid (6.7 mg, 0.0688 mmol), tetrakis(triphenylphosphine) palladium (2.4 mg, 0.00212 mmol) and CS2CO3 (41 mg, 0.127 mmol) were added. The reaction mixture was heated at 1100C for overnight. Then it was filtered and washed with methanol. The filtrate was concentrated and purified by Prep.HPLC to give yellowish oil as product. (12mg, 49% yield). MS m/z 470 (MH+). Example 79 Coupling Reaction: Example G
Figure imgf000149_0002
To a solution of A (20 mg, 0.0423 mmol) in DMF (1 mL), 3-furan boronic acid (6.2 mg, 0.055 mmol), tetrakis(triphenylphosphine) palladium (2.4 mg, 0.002115 mmol) and 2M Na2CO3 solution (0.064 mL, 0.127 mmol) were added. The reaction mixture was heated at 1100C for 2 days. Then it was filtered and washed with methanol. The filtrate was concentrated and purified by Prep.HPLC to give yellowish oil as product. (7.0 mg, 29% yield). Example 80 Coupling Reaction: Example H
Figure imgf000150_0001
To a solution of A (20 mg, 0.0423 mmol) in DMF (2 mL), 2- thiopheneboronic acid (7.0 mg, 0.055 mmol), tetrakis(triphenylphosphine) palladium (2.4 mg, 0.00212 mmol) and barium hydroxide (40 mg, 0.127 mmol) were added. The reaction mixture was heated at 1500C in Smith microwave reactor for 30 min. Then it was filtered and washed with methanol. The filtrate was concentrated and purified by Prep.HPLC to give brownish oil as product. (13.0 mg, 52% yield) 1H NMR (CD3OD, 400 MHz) δ 1.03 (s, 9 H), 2.18-2.25 (m, 2 H)3 2.93 (m, 1 H), 3.55 (s, 3 H), 3.83 (m, 1 H), 3.98 (m, 1 H), 4.34 (s, 1 H), 4.38 (m, 2 H), 4.58 (dd, J=8.05, 5.14 Hz, 1 H), 6.63 (d, J=8.07 Hz, 1 H), 7.07 (dd, J=4.89, 3.67 Hz, 1 H), 7.33 (d, ./=7.34 Hz, 1 H), 7.42 (d, J=5.14 Hz, 1 H), 7.60-7.66 (m, 2 H). MS m/z 476 (MH+). Example 81
Using the above coupling examples (A-H) as a reference in the design of reaction conditions, the following intermediates could be prepared. Each of these proposed intermediates (Intermediates 73-80) could then be converted into
Compounds of formula (I) by employing the teachings described, and referenced, herein.
Figure imgf000151_0001
Example 82
Preparation of Intermediate 82
Figure imgf000151_0002
To a solution of (2S, 4R) Fmoc-4-amino-l-boc-pyrrolidine-2-carboxylic acid (400 mg, 0.884 mmol) in acetonitrile (15 mL), five drops of pyrrolidine was added. The reaction mixture was stirred at it for 3hr. Then it was concentrated and put on high vacuum to give crude 4-amino-l-boc-pyrrolidine-2-carboxylic acid. In another round-bottomed flask, a solution of Pd2dba3 (40 mg, 5% mol)and racemic-BINAP (56 mg, 10% mol) was stirred under nitrogen in degassed toluene (8 mL) at rt for Ih. Then 1-chloroisoquinoline (216 mg, 1.326 mmol) and sodium t-butoxide (340 mg, 3.536 mmol) were added and the reaction mixture was stirred for 30 min. Then 4- amino-l-boc-pyrrolidine-2-carboxylic acid was added and the reaction mixture was heated under reflux for Ih. Water was added to quench the reaction and the aqueous layer was separated and filtered through filter paper. It was then concentrated and purified by Prep. HPLC to give coupled product as TFA salt. (165 mg, 40% yield) 1H NMR (CD3OD5 400 MHz) δ 1.44 (m, 9H)5 2.51-2.74 (m, 2H), 3.64 (m, IH), 4.01 (m, IH), 4.49 (m, IH), 4.64 (m, IH), 7.30 (d, J=6.85 Hz, IH), 7.58 (d, J=6.85 Hz, IH), 7.79(m, IH), 7.91-7.99 (m, 2H), 8.56 (d, J=8.56 Hz, IH). MS m/z 358 (MH+). Intermediate 82 can be used to make compounds of formula (I).
Figure imgf000152_0001
Example 83
Preparation of Intermediate 83
Figure imgf000152_0002
Step 1 : The tosylate of the Boc proline intermediate (A) was prepared as described in the literature (Patchett, A. A.; Witkof, B. J. Am. Chem. Soc. 1957, 185-192) and was used without further purification.
To a slurry of NaH (76 mg, 1.90 mmol) in DMF (20 ml) was added 1-thionaphthol (0.29 mg, 1.80 mmol) and the mixture stirred for 30 minutes. A solution of the Boc proline tosylate (0.61 g, 1.80 mmol) was added and the mixture stirred for 12 h at 230C. The mixture was concentrated and the residue partitioned between EtOAc/H2O. The organic extracts are dried (MgSO4) and concentrated. The residue was purified by column chromatography (elution with 5% EtOAc/hexanes to 30% EtOAc/hexanes to give 261 mg (38%) of the product as a yellow oil. 1H NMR (CDCl3, 3:2 mixture of rotamers) δ 1.41 (s, 9H)5 1.44 (s, 9H), 2.25-2.29 (m, 2H), 3.69 (s, 3H), 3.35-3.42 (m, IH), 3.51-3.53 (m, IH), 3.80-3.86 (m, 2H), 4.38-4.39 (m, IH), 4.46-4.48 (m, IH), 7.41-7.46 (m, IH), 7.42-7-54 (m, IH), 7.57-7.59 (m, IH), 7.58 (d, J = 4 Hz, IH), 7.82-7.88 (m, 2H), 8.46 (d, J = 5Hz, IH); MS m/z 388 (M++l). Intermediate 83 can be used to make compounds of formula (I).
Figure imgf000153_0001
Example 84
Preparation of Intermediate 84
Figure imgf000153_0002
To a slurry of NaH (76 mg, 1.90 mmol) in DMF (20 mL) was added 2- thionaphthol (0.29 g, 1.80 mmol) and the mixture was stirred for 30 minutes. A solution of the tosylate (Example 451, Step 1) (0.61 g, 1.79 mmol) in DMF (2 ml) was added and the mixture was stirred for 12 h ar 230C. The mixture was concentrated, then partitioned between EtOAc/H2O. The organic layer was washed with saturated NaHCO3, dried (MgSO4) and concentrated. The residue was chromatographed with 5% EtOAc/hexanes followed by 30% EtOAc/hexanes to give 261 mg (38%) of the product as a clear oil. 1H NMR (DMSO-d6) δ 1.32 (s, 9H), 2.29-2.35 (m, 2H), 3.33-3.47 (m, 2H), 3.66 (s, 3H), 3.71-3.81 (m, IH), 4.29-4.32 (s, IH), 7.49-7.55 (m, 3H), 7.70-7.80 (m,lH), 7.81-7.97 (m, 3H); MS m/z 387 (M+l). Intermediate 84 can be used to make compounds of formula (I).
Figure imgf000154_0001
Example 85
Preparation of Intermediate 85
Figure imgf000154_0002
To slurry of the sodium hydride (0.91 g, 22.7 mmol) in THF( 50 mL) was added N-BOC-trans-4(R)-hydroxy-L-proline (2.5 g, 10.8 mmol) and the mixture stirred at 230C for 1 h. 2-Chloromethylnapthalene (1.9 g, 10.8 mmol) was added and the mixture stirred for 12 h at room temperature. The solvent was removed and the residue poured into water and washed with hexanes. The aqueous layer was acidified (I N HCl) and extracted with EtOAc. The EtOAc layer is separated, dried (MgSO4), and concentrated to give a light yellow residue. The oil was purified by flash chromatography with 1 :1 EtOAc/hexanes with 1% acetic acid added to give 1.56 g (39%) of the desired product as a thick oil. 1H NMR (DMSO-d6, 3:1 mixture of rotamers) δ 1.35, 1.37 (s, 9H, major and minor respectively), 1.92-2.02, 2.15-2.20 (m, 2H, major and minor respectively), 2.35-2.50 (m, 2H), 3.41-3.49 (m, 2H), 4.12-4.16, 4.20-4.21 (m, 2H), 4.65-4.68(m, 2H), 7.46-7.52 (m, 3H), 7.74-7.91 (m, 4H), (Acid OH not observed); MS m/z 394 (M++1+Na). Intermediate 85 can be used to make compounds of formula (I).
Figure imgf000155_0001
Example 86
Preparation of Intermediate 86
Scheme 1
Figure imgf000155_0003
Figure imgf000155_0002
Figure imgf000155_0004
Step 1 : To a solution of commercially available N-Boc-(4S)-(cis)-Hydroxyproline- OMe (200mgs, 0.82 mmole), triphenylphosphine (320mgs, 1.22 mmole) and 1- naphthol (176mgs, 1.22 mmole) in 2.5 mL tetrahydrofuran was added dropwise a solution of diethyldiazodicarboxylate (190μL, 1.22 mmole) in 1.0 mL THF over 10 minutes. After stirring for 5.5 days, the reaction was concentrated in vacuo. The crude yellow oil was chromatographed on a 20X40cM preparative TLC plate (Analtech SiO2) eluting with 6-1 hexanes-ethyl acetate to yield the desired product as a pale yellow oil (150mgs, 33%). 1H NMR (CDCl3, 500MHz) δ 1.44 (s, 9H) 2.33 (IH, m), 2.72 (IH5 m), 3.77 and 3.38 (2s, 3H, rotamers), 3.88 (dd, IH, J= 4.3, 12.4 Hz), 3.97 (bd, IH), 4.53 and 4.62 (2t, IH, J=7.8Hz, rotamers), 5.10 (bd, IH), 6.76 (t, IH, J=9.5 Hz)5 7.37 (m, IH)5 7.46 (m, 3H), 7.80 (d, IH5 J=7.7 Hz), 8.18 (m, IH); MS m/z 394 (M+Na)+ Step 2: To a stirred solution of Boc-(4R)-naphthal-l-oxo)-Pro-OEt (150mgs, 0.40 mmole) in 1.5mL THF and 0.5mL water was added lithium hydroxide (lOmgs). The solution was stirred for 21 hours at room temperature and then diluted wih 0.5N NaHCO3. The basic solution was extracted with ethyl acetate and then the aqueous layer was acidified to pH 2 with the dropwise addition of cone. HCl. This acidified layer was then extracted again with ethyl acetate. This second ethyl acetate layer was dried with magnesium sulfate, filtered and then concentrated in vacuo to yield Boc- (4R)-naphthal-l-oxo)-Pro-OH as pale-pink crystals (147mgs, 100%). 1H NMR (CDCl3, 500MHz) δ 1.47 and 1.48 (2s, 9H, rotamers), 2.40 and 2.52 (2m, IH)5 2.68 and 2.78 (2m, IH), 3.78-4.07 (m, 2H), 4.57 and 4.69 (2t, IH, J=7.6 and 8.0 Hz, rotamers), 5.12 (bd, IH)5 6.77 (dd, IH5 J=7.6, 21.2 Hz), 7.37 (m, IH), 7.46 (m, 3H), 7.81 (t, IH, J=5.8 Hz), 8.19 (m, IH); MS m/z 358 (M+H)+. Step 3: To a solution of Boc-((4R)-naphthal-l-oxo)-Pro-OH (147mgs, 0.41 mmole) and racemic (lR/2S)/(lS/2R)-l-amino-2-vinylcyclopropane carboxylic acid ethyl ester hydrochloride salt (79mgs, 0.41 mmole) in 2.8mL methylene chloride was added DIPEA (250μL, 1.44 mmole) and TBTU (158mgs, 0.49 mmole). The resulting solution was stirred under nitrogen for 20 hours and then diluted with 4OmL methylene chloride. The organic layer was washed with water, IN NaHCO3, IN HCl, water and brine. The solution was then dried with sodium sulfate and concentrated in vacuo. Purification by preparative TLC yielded two separate diastereomers, higher Rf diastereomer A (P2[Boc(4R)-(naphthal- 1 -oxo)proline]- Pl(IR, 2S Vinyl Acca)-OEt, 78 mgs, 38%) and lower Rf diastereomer B (P2[Boc(4R)-(naphthal-l-oxo)proline]-Pl(lS, 2R Vinyl Acca)-OEt591mgs, 45%) as off white solids: Diastereomer A: P2[Boc(4R)-(naρhthal-l-oxo)proline]-Pl(lR, 2S Vinyl Acca)-OEt: 1H NMR (CDCl3, 500MHz) δ 1.24 (t, 3H), 1.43 (s, 9H), 1.52 (m, IH), 1.84 (m, IH), 2.02 (m, IH)5 2.14 (m, IH), 2.81 (m, IH), 3.88 (m, 2H), 4.11 (q, IH, J=7.15), 4.19 (m, IH), 4.54 (m, IH), 5.15 (m, IH), 5.31 (dd, IH, J=17, 0.8 Hz), 5.77 (m, IH), 6.83 (m, IH), 7.36 (t, IH, J=7.8 Hz), 7.46 (m, 3H), 7.78 (d, IH, J=7.6 Hz), 8.14 (d, IH,
J=8.15Hz); MS m/z 495 (M+H)+.
Diastereomer B, Example 1OB: P2[Boc(4R)-(naphthal-l-oxo)proline]-Pl(lS, 2R
Vinyl Acca)-OEt: 1H NMR CdI-CHCl3, 500MHz) δ 1.24 (t, 3H), 1.42 (s, 9H), 1.85
(m, IH), 2.15 (q, IH5 J=8.9Hz), 2.40 (m, IH), 2.78 (m, IH), 3.78 (m, IH), 4.12 (m,
2H), 4.52 (m, IH), 5.15 (m, IH), 5.31 (m, IH), 5.79 (m, IH), 6.80 (m, IH), 7.35 (t,
IH, J=7.6 Hz), 7.46 Cm, 3H), 7.78 Cd, IH, J=7.6 Hz), 8.14 Cd, IH, J=8.10 Hz).
MS m/z 495 CM+H)+.
Intermediate 86 can be used to make compounds of formula (I).
Figure imgf000157_0001
Biological Studies
HCV NS3/4A protease complex enzyme assays and cell-based HCV replicon assays were utilized in the present disclosure, and were prepared, conducted and validated as follows: Generation of recombinant HCV NS3/4A protease complex
HCV NS3 protease complexes, derived from the BMS strain, H77 strain or J4L6S strain, were generated, as described below. These purified recombinant proteins were generated for use in a homogeneous assay (see below) to provide an indication of how effective compounds of the present disclosure would be in inhibiting HCV NS3 proteolytic activity.
Serum from an HCV-infected patient was obtained from Dr. T. Wright, San Francisco Hospital. An engineered full-length cDNA Ccompliment deoxyribonucleic acid) template of the HCV genome CBMS strain) was constructed from DNA fragments obtained by reverse transcription-PCR CRT-PCR) of serum RNA Cribonucleic acid) and using primers selected on the basis of homology between other genotype Ia strains. From the determination of the entire genome sequence, a genotype Ia was assigned to the HCV isolate according to the classification of Simmonds et al. (See P Simmonds, KA Rose, S Graham, SW Chan, F McOmish, BC Dow, EA Follett, PL Yap and H Marsden, J. Clin. Microbiol., 31(6), 1493-1503 (1993)). The amino acid sequence of the nonstructural region, NS2-5B, was shown to be >97% identical to HCV genotype Ia (H77) and 87% identical to genotype Ib (J4L6S). The infectious clones, H77 (Ia genotype) and J4L6S (Ib genotype) were obtained from R. Purcell (NIH) and the sequences are published in Genbank (AAB67036, see Yanagi, M., Purcell, R.H., Emerson, S.U. and Bukh, J. Proc. Natl. Acad. ScL U.S.A. 94(16), 8738-8743 (1997); AF054247, see Yanagi,M., St Claire.M., Shapiro, M., Emerson, S.U., Purcell, R.H. and Bukh, J, Virology 244 (1), 161-172. (1998)).
The H77 and J4L6S strains were used for production of recombinant NS 3/4 A protease complexes. DNA encoding the recombinant HCV NS3/4A protease complex (amino acids 1027 to 1711) for these strains were manipulated as described by P. Gallinari et al. (see Gallinari P, Paolini C, Brennan D, Nardi C, Steinkuhler C, De Francesco R. Biochemistry. 38(17):5620-32, (1999)). Briefly, a three-lysine solubilizing tail was added at the 3'-end of the NS4A coding region. The cysteine in the Pl position of the NS4A-NS4B cleavage site (amino acid 1711) was changed to a glycine to avoid the proteolytic cleavage of the lysine tag. Furthermore, a cysteine to serine mutation was introduced by PCR at amino acid position 1454 to prevent the autolytic cleavage in the NS 3 helicase domain. The variant DNA fragment was cloned in the pET21b bacterial expression vector (Novagen) and the NS3/4A complex was expressed in Escherichia, coli strain BL21 (DE3) (Invitrogen) following the protocol described by P. Gallinari et al. (see Gallinari P, Brennan D, Nardi C, Brunetti M, Tomei L, Steinkuhler C, De Francesco R., J Virol. 72(8): 6758- 69 (1998)) with modifications. Briefly, the NS3/4A protease complex expression was induced with 0.5 millimolar (mM) Isopropyl β-D-1-thiogalactopyranoside (IPTG) for 22 hours (h) at 200C. A typical fermentation (1 Liter (L)) yielded approximately 10 grams (g) of wet cell paste. The cells were resuspended in lysis buffer (10 mL/g) consisting of 25 mM iV-(2-Hydroxyethyl)Piperazine-iVl-(2-Ethane Sulfonic acid) (HEPES), pH 7.5, 20% glycerol, 500 mM Sodium Chloride (NaCl), 0.5% Triton X-100, 1 microgram/milliliter ("μg/mL") lysozyme, 5 mM Magnesium Chloride (MgCl2), 1 μg/ml Dnasel, 5mM β-Mercaptoethanol (βME), Protease inhibitor-Ethylenediamine Tetraacetic acid (EDTA) free (Roche), homogenized and incubated for 20 minutes (min) at 4°C. The homogenate was sonicated and clarified by ultra-centrifugation at 235000 g for 1 h at 4°C. Imidazole was added to the supernatant to a final concentration of 15 mM and the pH adjusted to 8.0. The crude protein extract was loaded on a Nickel-Nitrilotriacetic acid (Ni-NTA) column pre- equilibrated with buffer B (25 mM HEPES, pH 8.0, 20% glycerol, 500 mM NaCl, 0.5% Triton X-IOO5 15 mM imidazole, 5 mM βME). The sample was loaded at a flow rate of 1 mL/min. The column was washed with 15 column volumes of buffer C (same as buffer B except with 0.2% Triton X-100). The protein was eluted with 5 column volumes of buffer D (same as buffer C except with 200 mM Imidazole).
NS3/4A protease complex-containing fractions were pooled and loaded on a desalting column Superdex-S200 pre-equilibrated with buffer D (25 mM HEPES, pH 7.5, 20% glycerol, 300 mM NaCl, 0.2% Triton X-100, 10 mM βME). Sample was loaded at a flow rate of 1 mL/min. NS3/4A protease complex-containing fractions were pooled and concentrated to approximately 0.5 mg/ml. The purity of the
NS3/4A protease complexes, derived from the BMS, H77 and J4L6S strains, were judged to be greater than 90% by SDS-PAGE and mass spectrometry analyses. The enzyme was stored at -80 0C, thawed on ice and diluted prior to use in assay buffer. FRET peptide assay to monitor HCV NS3/4A proteolytic activty The purpose of this in vitro assay was to measure the inhibition of HCV NS3 protease complexes, derived from the BMS strain, H77 strain or J4L6S strain, as described above, by compounds of the present disclosure. This assay provides an indication of how effective compounds of the present disclosure would be in inhibiting HCV NS 3 proteolytic activity. In order to monitor HCV NS3/4A protease activity, an NS3/4A peptide substrate was used. The substrate was RET Sl (Resonance Energy Transfer Depsipeptide Substrate; AnaSpec, Inc. cat # 2299I)(FRET peptide), described by Taliani et al. in Anal. Biochem. 240(2):60-67 (1996). The sequence of this peptide is loosely based on the NS4A/NS4B natural cleavage site for the HCV NS3 protease except there is an ester linkage rather than an amide bond at the cleavage site. The peptide also contains a fluorescence donor, EDANS, near one end of the peptide and an acceptor, DABCYL, near the other end. The fluorescence of the peptide is quenched by intermolecular resonance energy transfer (RET) between the donor and the acceptor, but as the NS3 protease cleaves the peptide the products are released from RET quenching and the fluorescence of the donor becomes apparent.
The peptide substrate was incubated with one of the three recombinant NS3/4A protease complexes, in the absence or presence of a compound of the present disclosure. The inhibitory effects of a compound was determined by monitoring the formation of fluorescent reaction product in real time using a Cytofluor Series 4000. The reagents were as follow: HEPES and Glycerol (Ultrapure) were obtained from GIBCO-BRL. Dimethyl Sulfoxide (DMSO) was obtained from Sigma, β- Mercaptoethanol was obtained from Bio Rad.
Assay buffer: 50 mM HEPES, pH 7.5; 0.15 MNaCl; 0.1% Triton; 15% Glycerol; 10 mM βME. Substrate: 2 μM final concentration (from a 2 mM stock solution in DMSO stored at -200C). HCV NS3/4A protease type Ia (Ib), 2-3 nM final concentration (from a 5 μM stock solution in 25 mM HEPES, pH 7.5, 20% glycerol, 300 mM NaCl, 0.2% Triton-XIOO, 10 mM βME). For compounds with potencies approaching the assay limit, the assay was made more sensitive by adding 50 μg/ml Bovine Serum Albumin (Sigma) to the assay buffer and reducing the end protease concentration to 300 pM.
The assay was performed in a 96-well polystyrene black plate from Falcon. Each well contained 25 μl NS3/4A protease complex in assay buffer, 50 μl of a compound of the present disclosure in 10% DMSO/assay buffer and 25 μl substrate in assay buffer. A control (no compound) was also prepared on the same assay plate. The enzyme complex was mixed with compound or control solution for 1 min before initiating the enzymatic reaction by the addition of substrate. The assay plate was read immediately using the Cytofluor Series 4000 (Perspective Biosystems). The instrument was set to read an emission of 340 nm and excitation of 490 nm at 25°C. Reactions were generally followed for approximately 15 min.
The percent inhibition was calculated with the following equation:
100-[(δFinh/δFcon)xl00] where 5F is the change in fluorescence over the linear range of the curve. A nonlinear curve fit was applied to the inhibition-concentration data, and the 50% effective concentration (ICs0) was calculated by the use of Excel XLfit software using the equation, y=A+((B-A)/(l+((C/x)AD))).
All of the compounds tested were found to inhibit the activity of the NS3/4A protease complex with IC50's of 0.55 μM or less. Further, compounds of the present disclosure, which were tested against more than one type of NS3/4A complex, were found to have similar inhibitory properties though the compounds uniformly demonstrated greater potency against the Ib strains as compared to the Ia strains.
Specificity Assays
The specificity assays were performed to demonstrate the in vitro selectivity of the compounds of the present disclosure in inhibiting HCV NS3/4A protease complex as compared to other serine or cysteine proteases.
The specificities of compounds of the present disclosure were determined against a variety of serine proteases: human neutrophil elastase (EQvTE), porcine pancreatic elastase (PPE) and human pancreatic chymotrypsin and one cysteine protease: human liver cathepsin B. In all cases, a 96-well plate format protocol using colorimetric p-nitroaniline (pNA) substrate or fluorometric Amino-Methyl-Coumarin (AMC) substrate, specific for each enzyme was used as described previously (PCT Patent Application No. WO 00/09543) with some modifications. All enzymes were purchased from Sigma or EMDbiosciences while the substrates were from Bachem.
Each pNA assay included a 2 h enzyme-inhibitor pre-incubation at room temperature followed by addition of substrate and hydrolysis to ~15% conversion as measured on a Spectramax Pro microplate reader. The cathepsin B assay was initiated by adding substrate to a 10 min enzyme-inhibitor pre-incubation at room temperature, and the assay plate measured immediately using the Cytofluor Series 4000. Compound concentrations varied from 100 to 0.4 μM depending on their potency.
The final conditions for each assay were as follows:
50 mM Tris(hydroxymethyl)aminomethane hydrochloride (Tris-HCl) pH 8, 0.5 M Sodium Sulfate (Na2SO4), 50 mM NaCl, 0.1 mM EDTA, 3% DMSO, 0.01% Tween- 20 with: 133 μM succ-AAA-pNA and 20 nM HNE or 8 nM PPE; 100 μM succ-AAPF-pNA and 250 pM Chymotrypsin. 100 mM NaHPO4 (Sodium Hydrogen Phosphate) pH 5.5, 3% DMSO, 1 mM TCEP (Tris(2-carboxyethyl)phosphine hydrochloride), 5 nM Cathepsin B (enzyme stock activated in buffer containing 20 mM TCEP before use), and 2 μM Z-FR-AMC diluted in H2O.
The percentage of inhibition was calculated using the formula: [ 1 -((UVinh-UVblank)/(UVcll-UVblank))] x 100
A non-linear curve fit was applied to the inhibition-concentration data, and the 50% effective concentration (IC50) was calculated by the use of Excel XLfit software.
Generation ofHCVReplicon An HCV replicon whole cell system was established as described by
Lohmann V, Korner F, Koch J, Herian U, Theilmann L, Bartenschlager R., Science 285(5424): 110-3 (1999). This system enabled us to evaluate the effects of our HCV Protease compounds on HCV RNA replication. Briefly, using the HCV strain Ib sequence described in the Lohmann paper (Assession number:AJ238799), an HCV cDNA was synthesized by Operon Technologies, Inc. (Alameda, CA), and the full- length replicon was then assembled in plasmid pGem9zf(+) (Promega, Madison, WI) using standard molecular biology techniques. The replicon consists of (i) the HCV 5' UTR fused to the first 12 amino acids of the capsid protein, (ii) the neomycin phosphotransferase gene (neo), (iii) the IRES from encephalomyocarditis virus (EMCV), and (iv) HCV NS3 to NS5B genes and the HCV 3 ' UTR. Plasmid DNAs were linearized with Seal and RNA transcripts were synthesized in vitro using the T7 MegaScript transcription kit (Ambion, Austin, TX) according to manufacturer's directions. In vitro transcripts of the cDNA were transfected into the human hepatoma cell line, HUH-7. Selection for cells constitutively expressing the HCV replicon was achieved in the presence of the selectable marker, neomycin (G418). Resulting cell lines were characterized for positive and negative strand RNA production and protein production over time.
HCV Replicon FRET Assay
The HCV replicon FRET assay was developed to monitor the inhibitory effects of compounds described in the disclosure on HCV viral replication. HUH-7 cells, constitutively expressing the HCV replicon, were grown in Dulbecco's Modified Eagle Media (DMEM) (Gibco-BRL) containing 10% Fetal calf serum (FCS) (Sigma) and 1 mg/ml G418 (Gibco-BRL). Cells were seeded the night before (1.5 x 104 cells/well) in 96-well tissue-culture sterile plates. Compound and no compound controls were prepared in DMEM containing 4% FCS5 1 : 100 Penicillin/Streptomysin (Gibco-BRL), 1:100 L-glutamine and 5% DMSO in the dilution plate (0.5% DMSO final concentration in the assay). Compound/DMSO mixes were added to the cells and incubated for 4 days at 37°C. After 4 days, cells were first assessed for cytotoxicity using alamar Blue (Trek Diagnotstic Systems) for a CC50 reading. The toxicity of compound (CC50) was determined by adding 1/10th volume of alamar Blue to the media incubating the cells. After 4 hours, the fluorescence signal from each well was read, with an excitation wavelength at 530 nm and an emission wavelength of 580 nm, using the Cytofluor Series 4000 (Perspective Biosystems). Plates were then rinsed thoroughly with Phosphate- Buffered Saline (PBS) (3 times 150 μl). The cells were lysed with 25 μl of a lysis assay reagent containing an HCV protease substrate (5X cell Luciferase cell culture lysis reagent (Promega #E153A) diluted to IX with distilled water, NaCl added to 150 mM final, the FRET peptide substrate (as described for the enzyme assay above) diluted to 10 μM final from a 2 mM stock in 100% DMSO. The HCV protease substrate. The plate was then placed into the Cytofluor 4000 instrument which had been set to 340 nm excitation/490 nm emission, automatic mode for 21 cycles and the plate read in a kinetic mode. EC50 determinations were carried out as described for the IC50 determinations.
HCV Repl icon, Luciferase Reporter Assay
As a secondary assay, EC50 determinations from the replicon FRET assay were confirmed in a replicon luciferase reporter assay. Utilization of a replicon luciferase reporter assay was first described by Krieger et al (Krieger N, Lohmann V, and Bartenschlager R, J. Virol. 75(10):4614-4624 (2001)). The replicon construct described for our FRET assay was modified by inserting cDNA encoding a humanized form of the Renilla luciferase gene and a linker sequence fused directly to the 3 '-end of the luciferase gene. This insert was introduced into the replicon construct using an Ascl restriction site located in core, directly upstream of the neomycin marker gene. The adaptive mutation at position 1179 (serine to isoleucine) was also introduced (Blight KJ, Kolykhalov, AA, Rice, CM, Science 290(5498): 1972-1974). A stable cell line constitutively expressing this HCV replicon construct was generated as described above. The luciferase reporter assay was set up as described for the HCV replicon FRET assay with the following modifications. Following 4 days in a 37°C/5% CO2 incubator, cells were analyzed for Renilla Luciferase activity using the Promega Dual-Glo Luciferase Assay System. Media (100 μl) was removed from each well containing cells. To the remaining 50 μl of media, 50 μl of Dual-Glo Luciferase Reagent was added, and plates rocked for 10 minutes to 2 hours at room temperature. Dual-Glo Stop & GIo Reagent (50 μl) was then added to each well, and plates were rocked again for an additional 10 minutes to 2 h at room temperature. Plates were read on a Packard TopCount NXT using a luminescence program.
The percentage inhibition was calculated using the formula below: % control = average luciferase signal in experimental wells (+ compound) average luciferase signal in DMSO control wells (- compound) The values were graphed and analyzed using XLfit to obtain the EC50 value. Representative compounds of the disclosure were assessed in the HCV enzyme assays, HCV replicon cell assay and/or in several of the outlined specificity assays. For example, Compound 3 was found to have an IC50 of 2.8 nanomolar (nM) against the NS3/4A BMS strain in the enzyme assay. Similar potency values were obtained with the published H77 (IC50 of 0.6 nM) and J4L6S (IC50 of 0.47 nM) strains. The EC50 value in the replicon FRET assay was 14.6 nM, and 5.5 nM in the replicon Luciferase assay.
In the specificity assays, the same compound was found to have the following activity: HLE > 25 μM; PPE > 100 μM; Chyrnotrypsin > lOOμM; Cathepsin B = 100 μM. These results indicate this family of compounds are highly specific for the NS3 protease and many of these members inhibit HCV replicon replication.
The compounds of the current disclosure were tested and found to have activities in the ranges as follow:
IC50 Activity Ranges (NS3/4A BMS Strain): A is > 1 micromolar (μM); B is 0.1-1 μM; C is < 0.1 μM EC50 Activity Range (for compounds tested): A is > 1 μM; B is 0.1-1 μM; C is < 0.1 μM
Note that by using the patent compound number shown in Table 1 the structures of compounds can be found herein.
In accordance with one embodiment of the present disclosure, the compounds have a biological activity (EC50) of 100 μM or less, and in another embodiment, 1 μM or less, and most preferably 0.1 μM or less.
Figure imgf000165_0002
Table 3 is a list of compounds that could be synthesized using the teachings described or referenced herein.
Figure imgf000165_0001
Figure imgf000165_0003
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Those skilled in the art will recognize that although the disclosure has been described above with respect to specific aspects, other aspects are intended to be within the scope of the claims which follow. AU documents referenced herein are hereby incorporated by reference as if set out in full.

Claims

CLAIMSWhat is claimed is:
1. A compound of formula (I)
Figure imgf000179_0001
or a pharmaceutically acceptable salt thereof, wherein
(a) R4 is H; Ci-β alkyl, or C3.7 cycloalkyl each optionally substituted with halo; alkoxy; -C(O)-R5; C(O)-N(R5)2; or C(O)-OR5, wherein each R5 is independently d.9 alkyl, optionally substituted with Ci .6 alkoxy, C3.7 cycloalkoxy, halo-Ci-6 alkoxy, cyario, halo, hydroxy, amino, Ci-6 alkylamino. di (C]-6) alkylamino, di (Cr6) alkylamide, carboxyl, (Cr6) carboxyester; C6_io aryl; C7-J4 alkylaryl; heterocylclyl; or C3-7 cycloalkyl optionally substituted with alkoxy, halo, or haloalkoxy;
(b) R6 is H, C1-6 alkyl, or C3-7 cycloalkyl;
(c) R3 and R3' are each independently H or methyl; (d) Q is a C3-9 saturated or unsaturated chain, optionally containing one to three heteroatoms independently selected from O or S(O)m; wherein m is 0, 1 or 2;
(e) W is OH, -O-R1, or -NH-SO2-R2; wherein R1 is Cj-6 alkyl, unsaturated C3.7 cycloalkyl, Q_i4 aryl or C7-I6 alkylaryl; and R2 is Cj-8 alkyl, C4.io alkylcycloalkyl, unsubstituted C3-7 cycloalkyl; or R2 is cyclopropyl or cyclobutyl optionally substituted with CM alkyl, C2-5 alkenyl, C7-I6 alkylaryl, alkoxy, alkoxyalkyl, C5.7 cycloalkyl, C5-7 eye loalkenyl, Ce-io alkylcycloalkyl, halo, haloalkyl, cyano, alkylcyano, haloalkoxy, or C(O)-X; wherein the C5.7 cycloalkyl, the Cs-7 cycloalkenyl, and the C6-Io alkylcycloalkyl are further optionally substituted with Ci-4 alkyl or hydroxy; and wherein X is selected from phenyl and -NHRX; wherein R* is selected from Ci-6 alkyl, Het, and C6-I0 aryl;
(f) X is O, S, SO, SO2, OCH2, CH2O or NH; (g) R' is Het, Cβ.io aryl or C7-I4 alkylaryl, each optionally substituted with one or more Ra groups; or R' is C3-9 cycloalkyl or Ci-7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; provided that when X is O5 then R' is other than
Figure imgf000180_0001
wherein Ra is H or Ra; Rx is selected from H, halo, Ci .6 alkyl, Ci -6 alkoxy, C3.β cycloalkyloxy, aryl, and Het; and Ry is selected from H, halo, CF3, Ci-β alkoxy, and C3-6 cycloalkyloxy; and
(h) each Ra is Ci .6 alkyl, C3-7 cycloalkyl, Ci-6 alkoxy, C3-7 cycloalkoxy, halo-Ci-β alkyl, CF3, mono-or di-halo-Ct-6 alkoxy, cyano, halo, thioalkyl, hydroxy, alkanoyl, NO2, SH, amino, Ci-6 alkylamino, di (Ci-6) alkylamino, di (Ci-6) alkylamide, carboxyl, (C\.β) carboxyester, Ci-^ alkylsulfone, Ci-ό alkylsulfonamide, di (Cue) alkyl(alkoxy)amine, C6-ioaryl, C7-I4 alkylaryl, or a 5-7 membered monocyclic heterocycle.
2. A compound of claim 1 wherein X is O.
3. A compound of claim 2 wherein R' is other than quinoline or isoquinoline.
4. A compound of claim 1 wherein X is selected from S, SO, SO2, OCH2, CH2O and NH.
5. A compound of formula (II)
Figure imgf000181_0001
or a pharmaceutically acceptable salt thereof, wherein
(g) R4 is -C(O)-R5, C(O)-NHR5 or C(O)-OR5; wherein R5 is C,_9 alkyl optionally substituted with Ci-6 alkoxy, cyano, halo;
(h) Q is a C5_7 saturated or unsaturated chain optionally containing one or two heteroatoms independently selected from O and S(O)m; wherein m is 0, 1 or 2; (i) W is OH or NH-SO2-R2, wherein R2 is Ci* alkyl, C4-io alkylcycloalkyl, unsubstituted C3-7 cyclo alkyl, or cyclopropyl or cyclobutyl optionally substituted with C i -4 alkyl or C7. i β alkylaryl;
(j) X is O3 S, OCH2, CH2O or NH;
(k) R' is Het, Ce-io aryl or C7-I4 alkylaryl, each optionally substituted with one or more Ra; or R' is C3-9 cycloalkyl or Ci-7 alkyl, each optionally substituted with halo, cyano, alkoxy, dialkylamino; provided that when X is O, then R' is other than
Figure imgf000181_0002
wherein Ra is H or Ra; Rx is selected from H, halo, Cue alkyl, C1^ alkoxy. C3-6 cycloalkyloxy, aryl, and Het; and Ry is selected from H, halo, CF3, C^ alkoxy, and C3-6 cycloalkyloxy; and (1) each Ra is C 1 -β alkyl, C3-7 cycloalkyl, C 1 -6 alkoxy. C3-7 cycloalkoxy, halo-C 1 ->6 alkyl, CF3, halo-Ci-6 alkoxy, cyano, halo, thioalkyl, hydroxy , amino, C!-6 alkylamino, di (Ci-6) alkylamino, di (Ci-6) alkylamide, carboxyl, (Crβ) c'arboxyester, C1-6 alkylsulfone, Ci.6 alkylsulfonamide, di (C^6) alkyl(alkoxy)amine, Cβ-io aryl, C7-K alkylaryl, or a 5-7 membered monocyclic heterocycle.
6. A compound of claim 5 wherein X is O.
7. A compound of claim 6 wherein R' is other than quinoline or isoquinoline.
8. A compound of claim 5 wherein R4 is C(O)-OR5; wherein R5 is C1^ alkyl.
9. A compound of claim 5 wherein Q is a C5-7 saturated or unsaturated chain optionally containing one oxygen atom.
10. A compound of claim 5 wherein W is NH-SO2-R2; wherein R2 is unsubstituted C3-7 cycloalkyl.
11. A composition comprising the compound of claim 1 and a pharmaceutically acceptable carrier.
12. The composition of claim 11 further comprising one or two additional compounds having anti-HCV activity.
13. The composition of claim 12 wherein at least one of the additional compounds is an interferon or ribavirin.
14. The composition of claim 13 wherein the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, and lymphoblastiod interferon tau.
15. The composition of claim 12 wherein at least one of the additional compounds is selected from ϊnterleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
16. The composition of claim 12 wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH.
17. A method of treating an HCV infection in a patient, comprising administering to the patient a therapeutically effective amount of the compound of claim 1.
18. The method of claim 17 wherein the method further comprises administering one or two additional compounds prior to, after, or simultaneously with the compound of claim 1.
19. The method of claim 18 wherein at least one of the additional compounds is an interferon or ribavirin.
20. The method of claim 19 wherein the interferon is selected from interferon alpha 2B, pegylated interferon alpha, consensus interferon, interferon alpha 2A, lymphoblastiod interferon tau.
21. The method of claim 18 wherein at least one of the additional compounds is selected from interleukin 2, interleukin 6, interleukin 12, a compound that enhances the development of a type 1 helper T cell response, interfering RNA, anti-sense RNA, Imiqimod, ribavirin, an inosine 5'-monophospate dehydrogenase inhibitor, amantadine, and rimantadine.
22. The method of claim 18 wherein at least one of the additional compounds is effective to inhibit the function of a target selected from HCV metalloprotease, HCV serine protease, HCV polymerase, HCV helicase, HCV NS4B protein, HCV entry, HCV assembly, HCV egress, HCV NS5A protein, and IMPDH.
PCT/US2006/042922 2005-11-03 2006-11-02 Hepatitis c virus inhibitors WO2007056120A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP06836865.3A EP1951743B1 (en) 2005-11-03 2006-11-02 Hepatitis c virus inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US73303905P 2005-11-03 2005-11-03
US60/733,039 2005-11-03
US73660605P 2005-11-14 2005-11-14
US60/736,606 2005-11-14

Publications (1)

Publication Number Publication Date
WO2007056120A1 true WO2007056120A1 (en) 2007-05-18

Family

ID=37762643

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/042922 WO2007056120A1 (en) 2005-11-03 2006-11-02 Hepatitis c virus inhibitors

Country Status (3)

Country Link
US (1) US7741281B2 (en)
EP (1) EP1951743B1 (en)
WO (1) WO2007056120A1 (en)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008057875A2 (en) * 2006-11-01 2008-05-15 Bristol-Myers Squibb Company Inhibitors of hepatitis c virus
WO2009005690A2 (en) * 2007-06-29 2009-01-08 Gilead Sciences, Inc. Antiviral compounds
WO2008137779A3 (en) * 2007-05-03 2009-01-08 Intermune Inc Novel macrocyclic inhibitors of hepatitis c virus replication
WO2009099596A2 (en) 2008-02-04 2009-08-13 Idenix Pharamaceuticals, Inc. Macrocyclic serine protease inhibitors
WO2009140500A1 (en) * 2008-05-15 2009-11-19 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2009142842A2 (en) * 2008-04-15 2009-11-26 Intermune, Inc. Novel macrocyclic inhibitors of hepatitis c virus replication
US7763584B2 (en) 2006-11-16 2010-07-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7772180B2 (en) 2006-11-09 2010-08-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2010101967A2 (en) 2009-03-04 2010-09-10 Idenix Pharmaceuticals, Inc. Phosphothiophene and phosphothiazole hcv polymerase inhibitors
WO2011017389A1 (en) 2009-08-05 2011-02-10 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors useful against viral infections, particularly hcv
US7888464B2 (en) 2006-11-16 2011-02-15 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7915291B2 (en) 2002-05-20 2011-03-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7935670B2 (en) 2006-07-11 2011-05-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2011063076A1 (en) 2009-11-19 2011-05-26 Itherx Pharmaceuticals, Inc. Methods of treating hepatitis c virus with oxoacetamide compounds
US7964560B2 (en) 2008-05-29 2011-06-21 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2011075615A1 (en) 2009-12-18 2011-06-23 Idenix Pharmaceuticals, Inc. 5,5-fused arylene or heteroarylene hepatitis c virus inhibitors
KR20110082516A (en) * 2008-09-04 2011-07-19 브리스톨-마이어스 스큅 컴퍼니 Process for synthesizing substituted isoquinolines
JP2011521965A (en) * 2008-05-29 2011-07-28 ブリストル−マイヤーズ スクイブ カンパニー Hepatitis C virus inhibitor
US8003604B2 (en) 2006-11-16 2011-08-23 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2012019299A1 (en) * 2010-08-11 2012-02-16 Boehringer Ingelheim International Gmbh Hepatitis c inhibitor compounds
US8163921B2 (en) 2008-04-16 2012-04-24 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8202996B2 (en) 2007-12-21 2012-06-19 Bristol-Myers Squibb Company Crystalline forms of N-(tert-butoxycarbonyl)-3-methyl-L-valyl-(4R)-4-((7-chloro-4-methoxy-1-isoquinolinyl)oxy)-N- ((1R,2S)-1-((cyclopropylsulfonyl)carbamoyl)-2-vinylcyclopropyl)-L-prolinamide
US8232246B2 (en) 2009-06-30 2012-07-31 Abbott Laboratories Anti-viral compounds
WO2012109398A1 (en) 2011-02-10 2012-08-16 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating hcv infections
WO2012135581A1 (en) 2011-03-31 2012-10-04 Idenix Pharmaceuticals, Inc. Methods for treating drug-resistant hepatitis c virus infection with a 5,5-fused arylene or heteroarylene hepatitis c virus inhibitor
JP2012532146A (en) * 2009-07-02 2012-12-13 ドクター・レディーズ・ラボラトリーズ・リミテッド Enzymes and methods for resolution of aminovinylcyclopropanecarboxylic acid derivatives
US8343477B2 (en) 2006-11-01 2013-01-01 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus
US8420596B2 (en) 2008-09-11 2013-04-16 Abbott Laboratories Macrocyclic hepatitis C serine protease inhibitors
US8691757B2 (en) 2011-06-15 2014-04-08 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2014071032A1 (en) * 2012-11-02 2014-05-08 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
US8937041B2 (en) 2010-12-30 2015-01-20 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
US8951964B2 (en) 2010-12-30 2015-02-10 Abbvie Inc. Phenanthridine macrocyclic hepatitis C serine protease inhibitors
US8957203B2 (en) 2011-05-05 2015-02-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2015042375A1 (en) 2013-09-20 2015-03-26 Idenix Pharmaceuticals, Inc. Hepatitis c virus inhibitors
US8993595B2 (en) 2009-04-08 2015-03-31 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors
WO2015134561A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Pharmaceutical compositions comprising a 5,5-fused heteroarylene flaviviridae inhibitor and their use for treating or preventing flaviviridae infection
WO2015134560A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Solid forms of a flaviviridae virus inhibitor compound and salts thereof
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
US9334279B2 (en) 2012-11-02 2016-05-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9409943B2 (en) 2012-11-05 2016-08-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9499550B2 (en) 2012-10-19 2016-11-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9580463B2 (en) 2013-03-07 2017-02-28 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9598433B2 (en) 2012-11-02 2017-03-21 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GEP20105124B (en) * 2005-07-25 2010-11-25 Array Biopharma Inc Novel macrocyclic inhibitors of hepatitis c virus replication
KR20080056295A (en) 2005-10-11 2008-06-20 인터뮨, 인크. Compounds and methods for inhibiting hepatitis c viral replication
US8268776B2 (en) 2006-06-06 2012-09-18 Enanta Pharmaceuticals, Inc. Macrocylic oximyl hepatitis C protease inhibitors
US9526769B2 (en) * 2006-06-06 2016-12-27 Enanta Pharmaceuticals, Inc. Macrocylic oximyl hepatitis C protease inhibitors
AR061629A1 (en) * 2006-06-26 2008-09-10 Enanta Pharm Inc QUINOXALINIL MACROCICLIC INHIBITORS OF SERINE PROTEASE OF HEPATITIS VIRUS C. PROCESS OF OBTAINING AND PHARMACEUTICAL COMPOSITIONS
RU2008152171A (en) * 2006-07-05 2010-08-10 Интермьюн, Инк. (Us) NEW HEPATITIS C VIRAL REPLICATION INHIBITORS
NZ574605A (en) 2006-07-13 2011-11-25 Achillion Pharmaceuticals Inc 4-amino-4-oxobutanoyl peptides as inhibitors of viral replication
WO2008019289A2 (en) 2006-08-04 2008-02-14 Enanta Pharmaceuticals, Inc. Tetrazolyl macrocyclic hepatitis c serine protease inhibitors
US20090035271A1 (en) * 2007-08-01 2009-02-05 Ying Sun Tetrazolyl macrocyclic hepatitis c serine protease inhibitors
US7718612B2 (en) * 2007-08-02 2010-05-18 Enanta Pharmaceuticals, Inc. Pyridazinonyl macrocyclic hepatitis C serine protease inhibitors
US20090098085A1 (en) * 2006-08-11 2009-04-16 Ying Sun Tetrazolyl acyclic hepatitis c serine protease inhibitors
US7687459B2 (en) * 2006-08-11 2010-03-30 Enanta Pharmaceuticals, Inc. Arylalkoxyl hepatitis C virus protease inhibitors
US7662779B2 (en) * 2006-08-11 2010-02-16 Enanta Pharmaceuticals, Inc. Triazolyl macrocyclic hepatitis C serine protease inhibitors
WO2008095999A1 (en) * 2007-02-08 2008-08-14 Tibotec Pharmaceuticals Ltd. Pyrimidine substituted macrocyclic hcv inhibitors
WO2008106130A2 (en) * 2007-02-26 2008-09-04 Achillion Pharmaceuticals, Inc. Tertiary amine substituted peptides useful as inhibitors of hcv replication
US7906513B2 (en) * 2007-04-26 2011-03-15 Enanta Pharmaceuticals, Inc. Hydrazide-containing hepatitis C serine protease inhibitors
US8377872B2 (en) 2007-04-26 2013-02-19 Enanta Pharmaceuticals, Inc. Cyclic P3 tripeptide hepatitis C serine protease inhibitors
US20080267917A1 (en) * 2007-04-26 2008-10-30 Deqiang Niu N-functionalized amides as hepatitis c serine protease inhibitors
AP2009005057A0 (en) 2007-05-10 2009-12-31 Array Biopharma Inc Novel peptide inhibitors of hepatitis c virus replication
WO2009070689A1 (en) * 2007-11-29 2009-06-04 Enanta Pharmaceuticals, Inc. Bicyclic, c5-substituted proline derivatives as inhibitors of the hepatitis c virus ns3 protease
US8263549B2 (en) * 2007-11-29 2012-09-11 Enanta Pharmaceuticals, Inc. C5-substituted, proline-derived, macrocyclic hepatitis C serine protease inhibitors
WO2009073719A1 (en) 2007-12-05 2009-06-11 Enanta Pharmaceuticals, Inc. Quinoxalinyl derivatives
WO2009073713A1 (en) * 2007-12-05 2009-06-11 Enanta Pharmaceuticals, Inc. Oximyl macrocyclic derivatives
CA2708047A1 (en) * 2007-12-06 2009-06-11 Enanta Pharmaceuticals, Inc. Process for making macrocyclic oximyl hepatitis c protease inhibitors
AU2008340261C1 (en) * 2007-12-21 2015-12-10 Celgene Avilomics Research, Inc. HCV protease inhibitors and uses thereof
US9163061B2 (en) 2007-12-21 2015-10-20 Celgene Avilomics Research, Inc. HCV protease inhibitors and uses thereof
US8309685B2 (en) * 2007-12-21 2012-11-13 Celgene Avilomics Research, Inc. HCV protease inhibitors and uses thereof
US8293705B2 (en) 2007-12-21 2012-10-23 Avila Therapeutics, Inc. HCV protease inhibitors and uses thereof
US8044087B2 (en) * 2008-09-29 2011-10-25 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
BRPI0922913A2 (en) * 2008-12-10 2015-08-18 Achillion Pharmaceuticals Inc Cyclic 4-amino-4-oxobutanoyl peptide analogs viral replication inhibitors
EP2364309B1 (en) 2008-12-10 2014-10-01 Achillion Pharmaceuticals, Inc. New 4-amino-4-oxobutanoyl peptides as inhibitors of viral replication
US8102720B2 (en) * 2009-02-02 2012-01-24 Qualcomm Incorporated System and method of pulse generation
AR075584A1 (en) * 2009-02-27 2011-04-20 Intermune Inc THERAPEUTIC COMPOSITIONS THAT INCLUDE beta-D-2'-DESOXI-2'-FLUORO-2'-C-METHYLYCTIDINE AND A CARDIEX ISOINDOL ACID DERIVATIVE AND ITS USES. COMPOUND.
CA2761650C (en) 2009-05-13 2015-05-26 Enanta Pharmaceuticals, Inc. Macrocyclic compounds as hepatitis c virus inhibitors
WO2011014882A1 (en) 2009-07-31 2011-02-03 Medtronic, Inc. CONTINUOUS SUBCUTANEOUS ADMINISTRATION OF INTERFERON-α TO HEPATITIS C INFECTED PATIENTS
TW201119667A (en) * 2009-10-19 2011-06-16 Enanta Pharm Inc Bismacrocyclic compounds as hepatitis C virus inhibitors
CA2822357A1 (en) 2010-12-22 2012-06-28 Abbvie Inc. Hepatitis c inhibitors and uses thereof
US20140100364A1 (en) 2012-10-08 2014-04-10 Abbvie Inc. Compounds Useful For Making HCV Protease Inhibitors
JP2016515139A (en) 2013-03-14 2016-05-26 アキリオン ファーマシューティカルズ,インコーポレーテッド A new method for producing buckwheatprevir
WO2014145507A1 (en) 2013-03-15 2014-09-18 Achillion Pharmaceuticals, Inc. A process for making a 4-amino-4-oxobutanoyl peptide cyclic analogue, an inhibitor of viral replication, and intermediates thereof
EP2970192A1 (en) 2013-03-15 2016-01-20 Achillion Pharmaceuticals, Inc. Sovaprevir polymorphs and methods of manufacture thereof
WO2014145600A1 (en) 2013-03-15 2014-09-18 Achillion Pharmaceuticals, Inc. Ach-0142684 sodium salt polymorphs, composition including the same, and method of manufacture thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003099274A1 (en) 2002-05-20 2003-12-04 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2004093798A2 (en) 2003-04-18 2004-11-04 Enanta Pharmaceuticals, Inc. Quinoxalinyl macrocyclic hepatitis c serine protease inhibitors
WO2005070955A1 (en) 2004-01-21 2005-08-04 Boehringer Ingelheim International Gmbh Macrocyclic peptides active against the hepatitis c virus
WO2006073145A1 (en) * 2005-01-04 2006-07-13 Calpis Co., Ltd Method of producing lactic acid bacterium having antiallergic effect
WO2007015824A2 (en) * 2005-07-25 2007-02-08 Intermune, Inc. Novel macrocyclic inhibitors of hepatitis c virus replication

Family Cites Families (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3719655A (en) * 1969-12-05 1973-03-06 Lilly Co Eli Process for the crystallization of the ammonium and alkali metal salts in insulin
DE3327928A1 (en) * 1983-08-03 1985-02-21 Hoechst Ag, 6230 Frankfurt METHOD FOR PRODUCING INSULIN DERIVATIVES
US5506203C1 (en) * 1993-06-24 2001-02-06 Astra Ab Systemic administration of a therapeutic preparation
BR9407508A (en) * 1993-09-17 1997-01-07 Novo Nordisk As Insulin derivative pharmaceutical composition and process for the treatment of diabetes in a patient in need of this treatment
US5700904A (en) * 1995-06-07 1997-12-23 Eli Lilly And Company Preparation of an acylated protein powder
AP1019A (en) 1996-10-18 2001-10-16 Vertex Pharma Inhibitors of serinre proteases, particularly hepatitis C virus NS3 protease.
US5898067A (en) * 1997-02-07 1999-04-27 Novo Nordisk A/S Crystallization of proteins
IL134232A0 (en) 1997-08-11 2001-04-30 Boehringer Ingelheim Ca Ltd Hepatitis c inhibitor peptides
HUP0100100A3 (en) 1997-08-11 2001-12-28 Boehringer Ingelheim Ca Ltd Hepatitis c inhibitor peptide analogues, pharmaceutical compositions comprising thereof and their use
US6323180B1 (en) 1998-08-10 2001-11-27 Boehringer Ingelheim (Canada) Ltd Hepatitis C inhibitor tri-peptides
AR022061A1 (en) 1998-08-10 2002-09-04 Boehringer Ingelheim Ca Ltd INHIBITING PEPTIDES OF HEPATITIS C, A PHARMACEUTICAL COMPOSITION CONTAINING THEM, THE USE OF THE SAME TO PREPARE A PHARMACEUTICAL COMPOSITION, THE USE OF AN INTERMEDIATE PRODUCT FOR THE PREPARATION OF THESE PEPTIDES AND A PROCEDURE FOR THE PREPARATION OF ANOGRAPH .
UA74546C2 (en) 1999-04-06 2006-01-16 Boehringer Ingelheim Ca Ltd Macrocyclic peptides having activity relative to hepatitis c virus, a pharmaceutical composition and use of the pharmaceutical composition
WO2002060926A2 (en) 2000-11-20 2002-08-08 Bristol-Myers Squibb Company Hepatitis c tripeptide inhibitors
US6867185B2 (en) 2001-12-20 2005-03-15 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus
CA2369711A1 (en) 2002-01-30 2003-07-30 Boehringer Ingelheim (Canada) Ltd. Macrocyclic peptides active against the hepatitis c virus
CA2370396A1 (en) 2002-02-01 2003-08-01 Boehringer Ingelheim (Canada) Ltd. Hepatitis c inhibitor tri-peptides
CA2369970A1 (en) 2002-02-01 2003-08-01 Boehringer Ingelheim (Canada) Ltd. Hepatitis c inhibitor tri-peptides
US6828301B2 (en) 2002-02-07 2004-12-07 Boehringer Ingelheim International Gmbh Pharmaceutical compositions for hepatitis C viral protease inhibitors
DE60334205D1 (en) 2002-05-20 2010-10-28 Bristol Myers Squibb Co Heterocyclische sulfonamid-hepatitis-c-virus-hemmer
US20060199773A1 (en) * 2002-05-20 2006-09-07 Sausker Justin B Crystalline forms of (1R,2S)-N-[(1,1-dimethylethoxy)carbonyl]-3-methyl-L-valyl-(4R)-4-[(6-methoxy-1-isoquinolinyl)oxy]-L-prolyl-1-amino-N-(cyclopropylsulfonyl)-2-ethenyl-cyclopropanecarboxamide, monopotassium salt
PL213029B1 (en) 2002-05-20 2012-12-31 Bristol Myers Squibb Co Substituted cycloalkyl p1' hepatitis c virus inhibitors
PL215228B1 (en) 2002-05-20 2013-11-29 Bristol Myers Squibb Co Hepatitis c virus inhibitors
US20040033959A1 (en) 2002-07-19 2004-02-19 Boehringer Ingelheim Pharmaceuticals, Inc. Pharmaceutical compositions for hepatitis C viral protease inhibitors
US20050075279A1 (en) 2002-10-25 2005-04-07 Boehringer Ingelheim International Gmbh Macrocyclic peptides active against the hepatitis C virus
US7601709B2 (en) 2003-02-07 2009-10-13 Enanta Pharmaceuticals, Inc. Macrocyclic hepatitis C serine protease inhibitors
EP1590442A4 (en) 2003-02-07 2007-07-18 Enanta Pharm Inc Macrocyclic hepatitis c serine protease inhibitors
ES2354282T3 (en) 2003-03-05 2011-03-11 Boehringer Ingelheim International Gmbh PEPTIDE ANALOGS INHIBITORS OF HEPATITIS C.
CA2516016C (en) 2003-03-05 2012-05-29 Boehringer Ingelheim International Gmbh Hepatitis c inhibiting compounds
KR20050108420A (en) 2003-04-02 2005-11-16 베링거 인겔하임 인터내셔날 게엠베하 Pharmaceutical compositions for hepatitis c viral protease inhibitors
ES2320771T3 (en) * 2003-04-16 2009-05-28 Bristol-Myers Squibb Company PEPTIDIC INHIBITORS OF MACROCICLIC ISOQUINOLINE VIRUS OF HEPATITIS C.
BRPI0410456B8 (en) 2003-05-21 2021-05-25 Boehringer Ingelheim Int hepatitis c inhibitor compounds, pharmaceutical composition, use thereof, as well as an article of manufacture
WO2004113365A2 (en) 2003-06-05 2004-12-29 Enanta Pharmaceuticals, Inc. Hepatitis c serine protease tri-peptide inhibitors
US7125845B2 (en) 2003-07-03 2006-10-24 Enanta Pharmaceuticals, Inc. Aza-peptide macrocyclic hepatitis C serine protease inhibitors
CN1856502A (en) 2003-09-22 2006-11-01 贝林格尔.英格海姆国际有限公司 Macrocyclic peptides active against the hepatitis c virus
RU2006115558A (en) 2003-10-10 2007-11-20 Вертекс Фармасьютикалз Инкорпорейтед (Us) SERINE PROTEASES INHIBITORS, ESPECIALLY HCV NS3-NS4A PROTEASES
CA2540858C (en) 2003-10-14 2009-12-08 Intermune, Inc. Macrocyclic carboxylic acids and acylsulfonamides as inhibitors of hcv replication
US7132504B2 (en) 2003-11-12 2006-11-07 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7135462B2 (en) 2003-11-20 2006-11-14 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7309708B2 (en) 2003-11-20 2007-12-18 Birstol-Myers Squibb Company Hepatitis C virus inhibitors
JP2007532479A (en) 2003-11-20 2007-11-15 シェーリング コーポレイション Depeptideization inhibitor of hepatitis C virus NS3 protease
JP4902361B2 (en) 2004-01-30 2012-03-21 メディヴィル・アクチエボラーグ HCVNS-3 serine protease inhibitor
EA012389B1 (en) 2004-03-30 2009-10-30 Интермун, Инк. Macrocyclic compounds as inhibitors of viral replication
WO2006016930A2 (en) 2004-05-14 2006-02-16 Intermune, Inc. Methods for treating hcv infection
CA2568008C (en) 2004-05-25 2014-01-28 Boehringer Ingelheim International Gmbh Process for preparing acyclic hcv protease inhibitors
JP4914348B2 (en) 2004-06-28 2012-04-11 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Hepatitis C inhibitor peptide analog
PL1778702T3 (en) 2004-07-16 2011-12-30 Gilead Sciences Inc Antiviral compounds
ATE512971T1 (en) 2004-07-20 2011-07-15 Boehringer Ingelheim Int PEPTIDE ANALOGUES AS HEPATITIS C INHIBITORS
UY29016A1 (en) 2004-07-20 2006-02-24 Boehringer Ingelheim Int ANALOGS OF INHIBITING DIPEPTIDES OF HEPATITIS C
WO2006026352A1 (en) 2004-08-27 2006-03-09 Schering Corporation Acylsulfonamide compounds as inhibitors of hepatitis c virus ns3 serine protease
US7375218B2 (en) 2004-09-17 2008-05-20 Boehringer Ingelheim International Gmbh Process for preparing macrocyclic HCV protease inhibitors
WO2007001406A2 (en) 2004-10-05 2007-01-04 Chiron Corporation Aryl-containing macrocyclic compounds
EP1805187A1 (en) * 2004-10-21 2007-07-11 Pfizer, Inc. Inhibitors of hepatitis c virus protease, and compositions and treatments using the same
US7323447B2 (en) 2005-02-08 2008-01-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
CA2600367C (en) 2005-03-08 2014-08-12 Boehringer Ingelheim International Gmbh Process for preparing macrocyclic compounds
CA2606195C (en) 2005-05-02 2015-03-31 Merck And Co., Inc. Hcv ns3 protease inhibitors
US7592336B2 (en) 2005-05-10 2009-09-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US20060276407A1 (en) 2005-06-02 2006-12-07 Schering Corporation Methods of treating hepatitis C virus
WO2006130687A2 (en) 2005-06-02 2006-12-07 Schering Corporation Liver/plasma concentration ratio for dosing hepatitis c virus protease inhibitor
US20070004635A1 (en) 2005-06-02 2007-01-04 Schering Corporation Method of treating interferon non-responders using HCV protease inhibitor
WO2006130552A2 (en) 2005-06-02 2006-12-07 Schering Corporation Methods of treating hepatitis c virus
WO2006130686A2 (en) 2005-06-02 2006-12-07 Schering Corporation Hcv protease inhibitors in combination with food
US20060281689A1 (en) 2005-06-02 2006-12-14 Schering Corporation Method for modulating activity of HCV protease through use of a novel HCV protease inhibitor to reduce duration of treatment period
WO2006130666A2 (en) 2005-06-02 2006-12-07 Schering Corporation Medicaments and methods combining a hcv protease inhibitor and an akr competitor
US20060276405A1 (en) 2005-06-02 2006-12-07 Schering Corporation Methods for treating hepatitis C
US7601686B2 (en) 2005-07-11 2009-10-13 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
JP2009501732A (en) 2005-07-20 2009-01-22 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Hepatitis C inhibitor peptide analog
TWI387603B (en) 2005-07-20 2013-03-01 Merck Sharp & Dohme Hcv ns3 protease inhibitors
MY144895A (en) 2005-07-29 2011-11-30 Tibotec Pharm Ltd Macrocylic inhibitors of hepatitis c virus
EP1913015B1 (en) 2005-07-29 2013-12-11 Janssen R&D Ireland Macrocyclic inhibitors of hepatitis c virus
PE20070210A1 (en) 2005-07-29 2007-04-16 Tibotec Pharm Ltd MACROCYCLIC COMPOUNDS AS INHIBITORS OF HEPATITIS C VIRUS
BRPI0614242A2 (en) 2005-07-29 2011-03-15 Medivir Ab macrocyclic hepatitis c virus inhibitors, combination and pharmaceutical composition comprising them, as well as use and process for the preparation of said inhibitors
PE20070343A1 (en) 2005-07-29 2007-05-12 Medivir Ab MACRO CYCLIC INHIBITORS OF HEPATITIS C VIRUS
JO2768B1 (en) 2005-07-29 2014-03-15 تيبوتيك فارماسيوتيكالز ليمتد Macrocylic Inhibitors Hepatitis C Virus
AR057703A1 (en) 2005-07-29 2007-12-12 Tibotec Pharm Ltd MACROCYCLIC INHIBITORS OF HEPATITIS VIRUS C. PHARMACEUTICAL PREPARATION AND COMPOSITION PROCESS.
PE20070211A1 (en) 2005-07-29 2007-05-12 Medivir Ab MACROCYCLIC COMPOUNDS AS INHIBITORS OF HEPATITIS C VIRUS
ES2456617T3 (en) 2005-07-29 2014-04-23 Janssen R&D Ireland Hepatitis C virus macrocyclic inhibitors
JP2009502845A (en) 2005-07-29 2009-01-29 メディヴィル・アクチボラグ Macrocyclic inhibitor of hepatitis C virus
WO2007014925A1 (en) 2005-07-29 2007-02-08 Tibotec Pharmaceuticals Ltd. Macrocylic inhibitors of hepatitis c virus
JP2009503084A (en) 2005-08-01 2009-01-29 フェノミックス コーポレーション Hepatitis C serine protease inhibitor and use thereof
BRPI0614205A2 (en) 2005-08-01 2016-11-22 Merck & Co Inc compound, pharmaceutical composition and compound use

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003099274A1 (en) 2002-05-20 2003-12-04 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
WO2004093798A2 (en) 2003-04-18 2004-11-04 Enanta Pharmaceuticals, Inc. Quinoxalinyl macrocyclic hepatitis c serine protease inhibitors
WO2005070955A1 (en) 2004-01-21 2005-08-04 Boehringer Ingelheim International Gmbh Macrocyclic peptides active against the hepatitis c virus
WO2006073145A1 (en) * 2005-01-04 2006-07-13 Calpis Co., Ltd Method of producing lactic acid bacterium having antiallergic effect
WO2007015824A2 (en) * 2005-07-25 2007-02-08 Intermune, Inc. Novel macrocyclic inhibitors of hepatitis c virus replication

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
TSANTRIZOS ET AL., ANGEWANDTE CHEMIE INT. ED., vol. 42, 2003, pages 1356 - 1360

Cited By (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9636375B2 (en) 2002-05-20 2017-05-02 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9227940B2 (en) 2002-05-20 2016-01-05 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8889871B2 (en) 2002-05-20 2014-11-18 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7915291B2 (en) 2002-05-20 2011-03-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7935670B2 (en) 2006-07-11 2011-05-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2008057875A3 (en) * 2006-11-01 2008-07-31 Bristol Myers Squibb Co Inhibitors of hepatitis c virus
WO2008057875A2 (en) * 2006-11-01 2008-05-15 Bristol-Myers Squibb Company Inhibitors of hepatitis c virus
US8343477B2 (en) 2006-11-01 2013-01-01 Bristol-Myers Squibb Company Inhibitors of hepatitis C virus
US7772180B2 (en) 2006-11-09 2010-08-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7888464B2 (en) 2006-11-16 2011-02-15 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8003604B2 (en) 2006-11-16 2011-08-23 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7763584B2 (en) 2006-11-16 2010-07-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2008137779A3 (en) * 2007-05-03 2009-01-08 Intermune Inc Novel macrocyclic inhibitors of hepatitis c virus replication
EP2177523A1 (en) * 2007-05-03 2010-04-21 Intermune, Inc. Novel macrocyclic inhibitors of hepatitis c virus replication
WO2009005690A3 (en) * 2007-06-29 2009-03-05 Gilead Sciences Inc Antiviral compounds
WO2009005690A2 (en) * 2007-06-29 2009-01-08 Gilead Sciences, Inc. Antiviral compounds
US8202996B2 (en) 2007-12-21 2012-06-19 Bristol-Myers Squibb Company Crystalline forms of N-(tert-butoxycarbonyl)-3-methyl-L-valyl-(4R)-4-((7-chloro-4-methoxy-1-isoquinolinyl)oxy)-N- ((1R,2S)-1-((cyclopropylsulfonyl)carbamoyl)-2-vinylcyclopropyl)-L-prolinamide
WO2009099596A2 (en) 2008-02-04 2009-08-13 Idenix Pharamaceuticals, Inc. Macrocyclic serine protease inhibitors
WO2009142842A2 (en) * 2008-04-15 2009-11-26 Intermune, Inc. Novel macrocyclic inhibitors of hepatitis c virus replication
WO2009142842A3 (en) * 2008-04-15 2010-04-01 Intermune, Inc. Novel macrocyclic inhibitors of hepatitis c virus replication
US8163921B2 (en) 2008-04-16 2012-04-24 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
CN102099359A (en) * 2008-05-15 2011-06-15 百时美施贵宝公司 Hepatitis c virus inhibitors
WO2009140500A1 (en) * 2008-05-15 2009-11-19 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
US7964560B2 (en) 2008-05-29 2011-06-21 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
JP2011521965A (en) * 2008-05-29 2011-07-28 ブリストル−マイヤーズ スクイブ カンパニー Hepatitis C virus inhibitor
KR20110082516A (en) * 2008-09-04 2011-07-19 브리스톨-마이어스 스큅 컴퍼니 Process for synthesizing substituted isoquinolines
KR101648595B1 (en) 2008-09-04 2016-08-16 브리스톨-마이어스 스퀴브 홀딩스 아일랜드 Process for synthesizing substituted isoquinolines
US8877929B2 (en) 2008-09-04 2014-11-04 Bristol-Myers Squibb Company Process for synthesizing substituted isoquinolines
JP2012515711A (en) * 2008-09-04 2012-07-12 ブリストル−マイヤーズ スクイブ カンパニー Method for the synthesis of substituted isoquinolines
JP2014159459A (en) * 2008-09-04 2014-09-04 Bristol Myers Squibb Co Process for synthesizing substituted isoquinolines
US8420596B2 (en) 2008-09-11 2013-04-16 Abbott Laboratories Macrocyclic hepatitis C serine protease inhibitors
US8642538B2 (en) 2008-09-11 2014-02-04 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
US9309279B2 (en) 2008-09-11 2016-04-12 Abbvie Inc. Macrocyclic hepatitis C serine protease inhibitors
WO2010101967A2 (en) 2009-03-04 2010-09-10 Idenix Pharmaceuticals, Inc. Phosphothiophene and phosphothiazole hcv polymerase inhibitors
US8993595B2 (en) 2009-04-08 2015-03-31 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors
US8232246B2 (en) 2009-06-30 2012-07-31 Abbott Laboratories Anti-viral compounds
JP2012532146A (en) * 2009-07-02 2012-12-13 ドクター・レディーズ・ラボラトリーズ・リミテッド Enzymes and methods for resolution of aminovinylcyclopropanecarboxylic acid derivatives
WO2011017389A1 (en) 2009-08-05 2011-02-10 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors useful against viral infections, particularly hcv
US9284307B2 (en) 2009-08-05 2016-03-15 Idenix Pharmaceuticals Llc Macrocyclic serine protease inhibitors
WO2011063076A1 (en) 2009-11-19 2011-05-26 Itherx Pharmaceuticals, Inc. Methods of treating hepatitis c virus with oxoacetamide compounds
WO2011075615A1 (en) 2009-12-18 2011-06-23 Idenix Pharmaceuticals, Inc. 5,5-fused arylene or heteroarylene hepatitis c virus inhibitors
WO2012019299A1 (en) * 2010-08-11 2012-02-16 Boehringer Ingelheim International Gmbh Hepatitis c inhibitor compounds
US8951964B2 (en) 2010-12-30 2015-02-10 Abbvie Inc. Phenanthridine macrocyclic hepatitis C serine protease inhibitors
US8937041B2 (en) 2010-12-30 2015-01-20 Abbvie, Inc. Macrocyclic hepatitis C serine protease inhibitors
WO2012109398A1 (en) 2011-02-10 2012-08-16 Idenix Pharmaceuticals, Inc. Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating hcv infections
US9353100B2 (en) 2011-02-10 2016-05-31 Idenix Pharmaceuticals Llc Macrocyclic serine protease inhibitors, pharmaceutical compositions thereof, and their use for treating HCV infections
WO2012135581A1 (en) 2011-03-31 2012-10-04 Idenix Pharmaceuticals, Inc. Methods for treating drug-resistant hepatitis c virus infection with a 5,5-fused arylene or heteroarylene hepatitis c virus inhibitor
US9527885B2 (en) 2011-05-05 2016-12-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8957203B2 (en) 2011-05-05 2015-02-17 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US10201541B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
US8691757B2 (en) 2011-06-15 2014-04-08 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9499550B2 (en) 2012-10-19 2016-11-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9598433B2 (en) 2012-11-02 2017-03-21 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9334279B2 (en) 2012-11-02 2016-05-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9643999B2 (en) 2012-11-02 2017-05-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2014071032A1 (en) * 2012-11-02 2014-05-08 Bristol-Myers Squibb Company Hepatitis c virus inhibitors
US9409943B2 (en) 2012-11-05 2016-08-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US9580463B2 (en) 2013-03-07 2017-02-28 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2015042375A1 (en) 2013-09-20 2015-03-26 Idenix Pharmaceuticals, Inc. Hepatitis c virus inhibitors
US9744170B2 (en) 2014-01-03 2017-08-29 Abbvie Inc. Solid antiviral dosage forms
US10105365B2 (en) 2014-01-03 2018-10-23 Abbvie Inc. Solid antiviral dosage forms
US9333204B2 (en) 2014-01-03 2016-05-10 Abbvie Inc. Solid antiviral dosage forms
WO2015134560A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Solid forms of a flaviviridae virus inhibitor compound and salts thereof
WO2015134561A1 (en) 2014-03-05 2015-09-11 Idenix Pharmaceuticals, Inc. Pharmaceutical compositions comprising a 5,5-fused heteroarylene flaviviridae inhibitor and their use for treating or preventing flaviviridae infection

Also Published As

Publication number Publication date
US7741281B2 (en) 2010-06-22
EP1951743B1 (en) 2014-03-19
US20070099825A1 (en) 2007-05-03
EP1951743A1 (en) 2008-08-06

Similar Documents

Publication Publication Date Title
EP1951743B1 (en) Hepatitis c virus inhibitors
EP2089385B1 (en) Inhibitors of hepatitis c virus
EP2086994B1 (en) Inhibitors of hepatitis c virus
EP1687018B1 (en) Hepatitis c virus inhibitors
EP1684787B1 (en) Hepatitis c virus inhibitors
EP2086967A2 (en) Inhibitors of hepatitis c virus
EP1945641B1 (en) Hepatitis c virus inhibitors
EP1506172B1 (en) Hepatitis c virus inhibitors
EP1684745B1 (en) Hepatitis c virus inhibitors
EP2813500B1 (en) Intermediates for the preparation of hepatitis C virus inhibitors
US20040077551A1 (en) Substituted cycloalkyl P1&#39; hepatitis C virus inhibitors
WO2008008776A2 (en) Hepatitis c virus inhibitors
EP1904516A2 (en) Hepatitis c virus inhibitors
EP2089020A1 (en) Macrocyclic peptides as hepatitis c virus inhibitors
EP2086980A1 (en) Macrocyclic peptides as hepatitis c virus inhibitors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006836865

Country of ref document: EP