HK1200847B - Inhibitors of hcv ns5a - Google Patents
Inhibitors of hcv ns5a Download PDFInfo
- Publication number
- HK1200847B HK1200847B HK15101205.2A HK15101205A HK1200847B HK 1200847 B HK1200847 B HK 1200847B HK 15101205 A HK15101205 A HK 15101205A HK 1200847 B HK1200847 B HK 1200847B
- Authority
- HK
- Hong Kong
- Prior art keywords
- compound
- hcv
- compounds
- pyrrolidine
- carbonyl
- Prior art date
Links
Description
The present invention provides non-nucleoside compounds of formulas I-III and certain derivatives thereof, which are useful as inhibitors of the Hepatitis C Virus (HCV) NS5A protein, inhibitors of HCV replication, and for the treatment of hepatitis C infections.
Hepatitis C Virus (HCV) infection is a major health problem leading to chronic liver diseases such as cirrhosis and hepatocellular carcinoma in many infected individuals. Current treatments for HCV infection include immunotherapy with recombinant interferon-alpha, alone or in combination with the nucleoside analog ribavirin.
Many virally encoded enzymes are putative targets for therapeutic intervention, including metalloproteases (NS2-3), serine proteases (NS3, amino acid residues 1-180), helicases (NS3, full length), NS3 protease cofactors (NS4A), membrane proteins (NS4B), zinc metalloproteins (NS5A), and RNA-dependent RNA polymerase (NS 5B).
One established therapeutic intervention target is the HCV NS5A nonstructural protein. The non-structural protein NS5A is an essential component of viral replication and assembly. Mutations at or near the known phosphorylation site in NS5A can affect the ability to replicate at high levels in cell culture systems, suggesting that NS5A phosphorylation plays an important role in viral replication efficiency. Inhibitors of NS5A phosphorylation may result in reduced viral RNA replication.
There is a clear and long-felt need to develop effective therapeutic agents for the treatment of HCV infection. In particular, there is a need to develop compounds useful for treating HCV-infected patients and compounds that selectively inhibit HCV viral replication.
Summary of The Invention
The present application provides compounds of formulas I-III or pharmaceutically acceptable salts thereof
Wherein:
each A is independently selected from
Each R1And R2Independently selected from H, lower alkyl or aryl;
each R3Independently selected from H, lower alkyl OR C (═ O) OR4;
R4Is a lower alkyl group;
each X is independently selected from H and Cl; and is
Each Y is1And Y2Independently selected from H or F.
The present application provides a method of treating a Hepatitis C Virus (HCV) infection comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any one of formulas I-III.
The present application provides a composition comprising a compound of any one of formulas I-III and a pharmaceutically acceptable excipient.
Detailed description of the invention
Definition of
The phrase "an" entity as used herein refers to one or more of such entities; for example, a compound refers to one or more compounds or at least one compound. In this regard, the terms "a", "an", "one or more" and "at least one" are used interchangeably herein.
The phrase "as defined herein above" refers to the broadest definition of each group as provided in the summary of the invention or in the broadest claims. In all other embodiments provided below, substituents that may be present in each embodiment and which are not explicitly defined retain the broadest definition provided in the summary of the invention.
As used in this specification, the term "comprising" shall be interpreted as having an open-ended meaning, whether in the conjunctive phrase or in the claims. That is, the term should be construed as synonymous with "having at least" or "including at least". When used in the context of a method, the term "comprising" means that the method includes at least the recited steps, but may also include additional steps. The term "comprising" when used in the context of a compound or composition means that the compound or composition includes at least the recited features or components, but may also include other features or components.
As used herein, unless otherwise expressly specified, the word "or" is used in the "nonexclusive" sense of "and/or" rather than the "exclusive" sense of "one of the" or "one.
The term "independently" as used herein means that a variable can be used in any one instance without regard to whether or not variables having the same or different definitions are present in the same compound. Thus, in a compound where R "occurs twice and is defined as" independently carbon or nitrogen, "both R" may be both carbon, both R "may be both nitrogen, or one R" may be carbon and the other nitrogen.
When any variable occurs more than one time in any moiety or formula depicting and describing compounds used or claimed in the present invention, its definition on each occurrence is independent of its definition at every other occurrence. Furthermore, combinations of substituents and/or variables are permissible only if such compounds result in stable compounds.
The symbol "at the end of a bond" or the symbol "through a bond" each refers to the point of attachment of a functional group or other chemical moiety to the rest of the molecule that it is a part of. Thus, for example:
MeC(=O)OR4wherein
A bond drawn into a ring system (as opposed to a bond attached to a distinct vertex) indicates that the bond may be attached to any suitable ring atom.
The term "optional" or "optionally" as used herein means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally substituted" means that the optionally substituted moiety may incorporate a hydrogen atom or a substituent.
The phrase "optional bond" means that the bond may or may not be present, and the description includes single, double, or triple bonds. If a substituent is referred to as a "bond" or "absent," the atom to which the substituent is attached is directly attached.
The term "about" as used herein is intended to mean approximately, within a certain area, approximately, or about. When the term "about" is used in conjunction with a numerical range, it modifies that range by extending the boundary values upward and or downward from the stated values. Generally, as used herein, the term "about" modifies a numerical value by a deviation in value of 20% upward and downward from the stated value.
Certain compounds may exhibit tautomerism. Tautomeric compounds may exist in the form of two or more interconvertible substances. Prototropic tautomers result from the migration of a covalently bonded hydrogen atom between two atoms. Tautomers generally exist in equilibrium, and attempts to isolate a single tautomer often result in a mixture whose chemical and physical properties are consistent with those of the compound mixture. The position of equilibrium depends on the chemical characteristics within the molecule. For example, in many aliphatic aldehydes and ketones such as acetaldehyde, the keto form predominates; whereas in phenol the enol type predominates. Common prototropic tautomers include keto/enol (-C (═ O) -CH-D-C (-OH) ═ CH-), amide/imidic (-C (═ O) -NH-D-C (-OH) ═ N-) and amidine (-C (═ NR) -NH-D-C (-NHR) ═ N-) tautomers. The latter two are particularly common in heteroaryl and heterocyclic rings, and the present invention encompasses all tautomeric forms of the compounds.
Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Reference may be made to various methods and materials known to those skilled in the art. Standard references to explain The rationale for pharmacology include The Pharmacological Basis of therapeutics, 10 th edition, McGraw Hill Companies Inc., New York (2001), Goodman and Gilman. The invention may be practiced with any suitable materials and/or methods known to those skilled in the art. However, preferred materials and methods are described. Unless otherwise indicated, materials, reagents, and the like referred to in the following description and examples may be obtained from commercial sources.
The definitions described herein may be appended to form chemically relevant combinations such as "heteroalkylaryl," "haloalkylheteroaryl," "arylalkyl heterocyclyl," "alkylcarbonyl," "alkoxyalkyl," and the like. When the term "alkyl" is used as a suffix following another term, as in "phenylalkyl" or "hydroxyalkyl", it means an alkyl group, as defined above, substituted with one to two substituents selected from the other groups specifically identified. Thus, for example, "phenylalkyl" refers to an alkyl group having one to two phenyl substituents, and thus includes benzyl, phenylethyl, and biphenyl. "alkylaminoalkyl" is an alkyl group having one to two alkylamino substituents. "hydroxyalkyl" includes 2-hydroxyethyl, 2-hydroxypropyl, 1- (hydroxymethyl) -2-methylpropyl, 2-hydroxybutyl, 2, 3-dihydroxybutyl, 2- (hydroxymethyl), 3-hydroxypropyl and the like. Thus, as used herein, the term "hydroxyalkyl" is used to define a subset of heteroalkyl groups defined below. The term- (ar) alkyl refers to unsubstituted alkyl or aralkyl. The term (hetero) aryl refers to aryl or heteroaryl.
The term "spirocycloalkyl" as used herein means a spirocyclic cycloalkyl group, for example, spirocyclo [3.3] heptane. The term spiroheterocycloalkyl as used herein refers to a spirocyclic heterocycloalkyl group, for example, 2, 6-diazaspiro [3.3] heptane.
The term "acyl" as used herein refers to a group of formula-C (═ O) R, where R is hydrogen or lower alkyl as defined herein. The term "alkylcarbonyl" as used herein, denotes a group of formula C (═ O) R, wherein R is alkyl as defined herein. Term C1-6Acyl refers to a-C (═ O) R group containing 6 carbon atoms. The term "arylcarbonyl" as used herein refers to a group of formula C (═ O) R, wherein R is aryl; the term "benzoyl" as used herein refers to an "arylcarbonyl" group wherein R is phenyl.
The term "ester" as used herein denotes a group of formula-C (═ O) OR, where R is lower alkyl as defined herein.
The term "alkyl" as used herein denotes an unbranched or branched saturated monovalent hydrocarbon residue containing from 1 to 10 carbon atoms. The term "lower alkyl" denotes a straight or branched chain hydrocarbon residue comprising 1 to 6 carbon atoms. As used herein, "C1-10Alkyl "refers to an alkyl group consisting of 1 to 10 carbons. Examples of alkyl include, without limitation, lower alkyl groups including methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl or pentyl, isopentyl, neopentyl, hexyl, heptyl, and octyl.
When the term "alkyl" is used as a suffix following another term, as in "phenylalkyl" or "hydroxyalkyl", it means an alkyl group, as defined above, substituted with one to two substituents selected from the other groups specifically identified. Thus, for example, "phenylalkyl" represents a group R 'R "-, where R' is phenyl and R" is alkylene as defined herein, it being understood that the point of attachment of the phenylalkyl group will be on the alkylene. Examples of arylalkyl groups include, but are not limited to, benzyl, phenylethyl, 3-phenylpropyl. The term "arylalkyl" or "aralkyl" has a similar interpretation, except that R' is aryl. The terms "(hetero) arylalkyl" or "(hetero) aralkyl" have similar interpretation except that R' is optionally aryl or heteroaryl.
The term "haloalkyl" or "halo-lower alkyl" or "lower haloalkyl" refers to a straight or branched chain hydrocarbon residue comprising 1 to 6 carbon atoms in which one or more carbon atoms are substituted with one or more halogen atoms.
The term "alkylene" as used herein, unless otherwise specified, denotes a divalent saturated straight-chain hydrocarbon group of 1 to 10 carbon atoms (e.g., (CH)2)n) Or a branched saturated divalent hydrocarbon group of 2 to 10 carbon atoms (e.g., -CHMe-or-CH)2CH(i-Pr)CH2-). The open valences of the alkylene groups are not bound to the same atom, except for the methylene group. Examples of alkylene include, without limitation, methylene, ethylene, propylene, 2-methyl-propylene, 1-dimethyl-ethylene, butylene, 2-ethylbutylene.
The term "alkoxy" as used herein means an-O-alkyl group wherein alkyl is as defined above, such as methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, t-butoxy, pentyloxy, hexyloxy, including isomers thereof. "lower alkoxy" as used herein denotes alkoxy having "lower alkyl" as defined above. As used herein, "C1-10Alkoxy "means wherein alkyl is C1-10an-O-alkyl group of (a).
The term "PCy3"means being surrounded by three ringsPartially trisubstituted phosphines.
The term "haloalkoxy" or "halo-lower alkoxy" or "lower haloalkoxy" refers to lower alkoxy wherein one or more carbon atoms are substituted with one or more halogen atoms.
The term "hydroxyalkyl" as used herein denotes an alkyl group as defined herein wherein one to three hydrogen atoms on different carbon atoms are replaced by a hydroxyl group.
The terms "alkylsulfonyl" and "arylsulfonyl" as used herein refer to the formula-S (═ O)2R, wherein R is independently alkyl or aryl and alkyl and aryl are as defined herein. The term "heteroalkylsulfonyl" as used herein denotes a group of formula-S (═ O) where R is "heteroalkyl" as defined herein2The radical of R.
The terms "alkylsulfonylamino" and "arylsulfonylamino" as used herein refer to the formula-NR' S (═ O)2R, wherein R is independently alkyl or aryl, R' is hydrogen or C1-3Alkyl, and alkyl and aryl are as defined herein.
The term "cycloalkyl" as used herein refers to a saturated carbocyclic ring containing 3 to 8 carbon atoms, i.e., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl. As used herein, "C3-7Cycloalkyl "refers to cycloalkyl groups containing from 3 to 7 carbons in the carbocyclic ring.
The term carboxy-alkyl as used herein refers to an alkyl moiety wherein one hydrogen atom is replaced by a carboxy group, it being understood that the heteroalkyl group is attached through a carbon atom. The term "carboxy" refers to-CO2And (4) a H part.
The term "heteroaryl" or "heteroaromatic" as used herein means a monocyclic or bicyclic group of 5 to 12 ring atoms having at least one aromatic or partially unsaturated ring, each ring containing 4 to 8 atoms in admixture with one or more N, O or S heteroatoms, the remaining ring atoms being carbon, it being understood thatThe point of attachment of the heteroaryl group is on an aromatic or partially unsaturated ring. As is well known to those skilled in the art, heteroaryl rings have less aromatic character than their all-carbon analogs. Thus, for the purposes of the present invention, heteroaryl groups need only have some degree of aromatic character. Examples of heteroaryl moieties include monocyclic aromatic heterocycles having 5 to 6 ring atoms and 1 to 3 heteroatoms, including, without limitation, pyridyl, pyrimidinyl, pyrazinyl, and the like,Oxazinyl, pyrrolyl, pyrazolyl, imidazolyl,Azolyl, 4, 5-dihydro-Azolyl, 5, 6-dihydro-4H- [1,3]Azolyl radical, isoOxazole, thiazole, isothiazole, triazoline, thiadiazole and oxadixoline, which may be optionally substituted with one or more, preferably one or two, substituents selected from the group consisting of hydroxy, cyano, alkyl, alkoxy, thio, lower haloalkoxy, alkylthio, halo, lower haloalkyl, alkylsulfinyl, alkylsulfonyl, halogen, amino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl and dialkylaminoalkyl, nitro, alkoxycarbonyl and carbamoyl, alkylcarbamoyl, dialkylcarbamoyl, arylcarbamoyl, alkylcarbonylamino and arylcarbonylamino. Examples of bicyclic moieties include, without limitation, quinolinyl, isoquinolinyl, benzofuranyl, benzothienylAzole, benzisohOxazole, benzothiazole, naphthyridinyl, 5,6,7, 8-tetrahydro- [1,6]Naphthyridinyl and benzisothiazole. The bicyclic moiety may be optionally substituted on any one of the rings, but the point of attachment is on the ring containing the heteroatom.
As used herein, unless otherwise indicated, the term "heterocyclyl", "heterocycloalkyl" or "heterocycle" means one or more rings consisting of one or more rings of 3 to 8 atoms per ring, preferably one to two rings (including spiro ring systems), mixed with one or more ring heteroatoms (selected from N, O or S (O))0-2) Optionally independently substituted with one or more, preferably one or two, substituents selected from the group consisting of hydroxy, oxo, cyano, lower alkyl, lower alkoxy, lower haloalkoxy, alkylthio, halogen, lower haloalkyl, hydroxyalkyl, nitro, alkoxycarbonyl, amino, alkylamino, alkylsulfonyl, arylsulfonyl, alkylaminosulfonyl, arylaminosulfonyl, alkylsulfonylamino, arylsulfonylamino, alkylaminocarbonyl, arylaminocarbonyl, alkylcarbonylamino, arylcarbonylamino, and ionic forms thereof. Examples of heterocyclic groups include, but are not limited to, morpholinyl, piperazinyl, piperidinyl, azetidinyl, pyrrolidinyl, hexahydroazepinyltrienyl, oxetanyl, tetrahydrofuranyl, tetrahydrothienyl,oxazolidinyl, thiazolidinyl, isooxazolidinylOxazolidinyl, tetrahydropyranyl, thiomorpholinyl, quinuclidinyl, and imidazolinyl, as well as ionic forms thereof. Examples thereof may also be bicyclic rings, such as 3, 8-diaza-bicyclo [3.2.1]Octane, 2, 5-diaza-bicyclo [2.2.2]Octane or octahydro-pyrazino [2,1-c][1,4]And (3) an oxazine.
Inhibitors of HCV NS5A
The present application provides compounds of formulas I-III or pharmaceutically acceptable salts thereof
Wherein:
each A is independently selected from
Each R1And R2Independently selected from H, lower alkyl or aryl;
each R3Independently selected from H, lower alkyl OR C (═ O) OR4;
R4Is a lower alkyl group;
each X is independently selected from H and Cl; and
each Y is1And Y2Independently selected from H or F.
The present application provides compounds of formula I-III wherein A is biphenyl.
The present application provides compounds of formula I-III wherein X is H.
The present application provides compounds of formula I-III wherein X is Cl.
The present application provides compounds of formula I-III wherein X is H and A is biphenyl.
The present application provides compounds of formula I-III wherein X is Cl and A is biphenyl.
The application provides wherein each R2Compounds of formulae I-III that are H.
The application provides wherein each R3Is C (═ O) OCH3Compounds of formulae I-III.
The application provides wherein each R2Is H and each R3Is C (═ O) OCH3Compounds of formulae I-III.
Provided herein are those wherein X is H, A is biphenyl, each R2Is H, and each R3Is C (═ O) OCH3Compounds of formulae I-III.
The present application provides compounds of formula I-III wherein R is methyl.
Provided herein are those wherein X is H, A is biphenyl, each R2Is H, and each R3Is C (═ O) OCH3Compounds of formulae I-III.
The application provides wherein R1Compounds of formula I-III which are isopropyl.
The present application provides wherein two R1Compounds of formula III, both isopropyl.
The application provides one of R1A compound of formula III which is isopropyl and the other is phenyl.
The present application provides wherein two R1Compounds of formula III which are both phenyl.
Provided herein are those wherein X is H, A is biphenyl, each R2Is H, each R3Is C (═ O) OCH3And R is1Compounds of formula I-III which are isopropyl.
The present application provides wherein Y1Compounds of formulae I-III that are H.
Provided herein are those wherein X is H, A is biphenyl, each R2Is H, each R3Is C (═ O) OCH3,Y1Is H, and R1Compounds of formula I-III which are isopropyl.
The present application provides wherein Y2Compounds of formulae I-III that are H.
Provided herein are those wherein X is H, A is biphenyl, each R2Is H, each R3Is C (═ O) OCH3,Y1Is H, Y2Is H, and R1Compounds of formula I-III which are isopropyl.
The present application provides wherein Y1Compounds of formulae I-III are F.
Provided herein are those wherein X is H, A is biphenyl, each R2Is H, each R3Is C (═ O) OCH3,Y1Is F, and R1Compounds of formula I-III which are isopropyl.
Provided herein are those wherein X is H, A is biphenyl, each R2Is H, each R3Is C (═ O) OCH3,Y1Is F, Y2Is H, and R1Compounds of formula I-III which are isopropyl.
The present application provides wherein Y2Compounds of formulae I-III are F.
Provided herein are those wherein X is H, A is biphenyl, each R2Is H, each R3Is C (═ O) OCH3,Y1Is F, Y2Is F, and R1Compounds of formula I-III which are isopropyl.
The present application provides a compound selected from the group consisting of:
{ (2S,5S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ] indol-5-yl } -carbamic acid methyl ester;
{ (4S,7S) -4- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -6, 10-dioxo-octahydro-pyridazino [1,2-a ] [1,2] diazepin-7-yl } -carbamic acid methyl ester;
{ (2S,5S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -4, 4-difluoropyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ] indol-5-yl } -carbamic acid methyl ester; and
{ (2S,5S) -2- [ (S) -2- (4' - { 5-chloro-2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ] indol-5-yl } -carbamic acid methyl ester.
The present application provides a method of treating a Hepatitis C Virus (HCV) infection comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any one of formulas I-III.
The present application provides the above method further comprising administering an immune system modulator or antiviral agent that inhibits replication of HCV, or a combination thereof.
The present application provides the above method, wherein the immune system modulator is an interferon or a chemically derivatized interferon.
The present application provides the above methods, wherein the antiviral agent is selected from the group consisting of an HCV protease inhibitor, an HCV polymerase inhibitor, an HCV helicase inhibitor, an HCV primase inhibitor, an HCV fusion inhibitor, and combinations thereof.
The present application provides a method of inhibiting HCV replication in a cell comprising administering a compound of any one of formulas I-III.
The present application provides a composition comprising a compound of any one of formulas I-III and a pharmaceutically acceptable excipient.
The present application provides the use of a compound of any one of formulas I-III for the manufacture of a medicament for the treatment of HCV.
The present application provides a compound, composition or method as described herein.
Compound (I)
Examples of representative compounds encompassed by the present invention within the scope of the present invention are provided in the following table. These examples and preparations are provided below to enable those skilled in the art to more clearly understand and practice the present invention. They should not be construed as limiting the scope of the invention, which is merely an illustrative and representative example of the invention.
Nomenclature used herein is generally based on AUTONOMTMVersion 4.0, a Beilstein Institute computerized system for generating IUPAC systematic nomenclature. If there is inconsistency between the drawn structure and the structure given by the name, the drawn structure is taken as a weight. Further, if the stereochemistry of a structural formula or a portion of a structural formula is not indicated with, for example, bold or dashed lines, the structural formula or the portion of the structural formula is to be understood as encompassing all stereoisomers thereof.
Table I describes some examples of compounds of formulas I-III.
Table I.
Synthesis of
General procedure
Wherein A may be independently selected from
Each R1And R2May be independently selected from H, lower alkyl or aryl, each R3May be independently selected from H, lower alkyl OR C (═ O) OR4,R4May be lower alkyl, each X may be independently selected from H and Cl and each Y1And Y2Compounds of formulae I-III as described herein, which may be independently selected from H or F, may generally be synthesized according to the general schemes drawn and described below.
Compounds of formulae I-III, wherein the variable X on the imidazole may be Cl, the imidazole may be a chloro-imidazole moiety, a is independently biphenyl, quinolyl, naphthyl, quinazolinyl or quinoxalinyl or other biaryl moieties as previously defined, and B in structures 1 and 2 may be hexahydro-pyridazino [1,2-a ] [1,2] diazepino-6, 10-dione or 1,2,6, 7-tetrahydro-5H-azepino [3,2,1-hi ] indol-4-one or N-acylpyrrolidine, may be prepared from the corresponding imidazole 1, with, for example, the Journal of Medicinal Chemistry (1986),29(6), 1065-80; journal of the chemical Society, Perkin Transactions1, Organic and Bio-Organic Chemistry (1972-1999) (1983) (4), 809-11; eur.pat.appl. (1990) EP365030a 119900425; journal of heterocyclic Chemistry (1994),31(5), 1121-3; PCT int.appl. (2007), WO2007070433, under standard reaction conditions for the chlorination of imidazole derivatives. (scheme 1).
Scheme 2
The compounds of formula 1 can be prepared by coupling of amine 2 and carboxylic acid 3 by standard peptide bond formation reactions. The compounds of formula 2 can be readily prepared from the compounds of formula 4 by deprotection of the amine group. (scheme 3).
Scheme 3
The compound of formula 4 can be prepared by coupling the compounds of formulae 5 and 6, and the resulting compound can be heated in a solvent such as toluene in the presence of ammonium acetate (scheme 4).
Scheme 4
The compounds of formula 5 are readily available starting from proline derivatives. The compounds of formula 6 can be derived from the corresponding carboxylic acids, which can be prepared by the general reaction scheme shown (scheme 5).
Scheme 5
Compounds of formula 3 wherein "B" is a tricyclic hexahydro-pyridazino [1,2-a ] [1,2] diazepin-6, 10-dione can be prepared starting from commercially available FMOC-protected intermediate 7 by preparing intermediates of formula 8 (scheme 6). Intermediate 8 can be coupled with a readily available suitably protected proline derivative to provide intermediate 3.
Scheme 6
Compounds of formula 3 wherein "B" is a bicyclic 1,2,6, 7-tetrahydro-5H-azepino [3,2,1-hi ] indol-4-one can be prepared starting from 9 (Attwood, M.R et al, Journal of the Chemical Society, PerkinTransactions1: Organic and Bio-Organic Chemistry (1972. sup. 1999)1986,1011-19) by converting it to derivative 10, which derivative 10 can be converted to a compound of formula 3 by standard reaction sequences involving suitably protected proline derivatives (scheme 7). The compounds of formula 3 wherein "B" is N-acylpyrrolidine are readily obtainable from the corresponding proline derivatives.
Scheme 7
Pharmaceutical compositions and administration
Pharmaceutical compositions of the subject compounds were prepared for administration by various routes as described in this example.
Composition for oral administration (A)
Mixing the above materials, and packaging into capsules, each containing about 100 mg; one capsule approximates one total daily dose.
Composition for oral administration (B)
The ingredients are combined and granulated with a solvent (e.g., methanol). The formulation is then dried and tableted with a suitable tabletting machine (containing about 20mg of active compound).
Composition for oral administration (C)
These ingredients are mixed and made into a suspension for oral administration.
Parenteral preparation (D)
The active ingredient is dissolved in a portion of the water for injection. Sufficient sodium chloride is then added with stirring to make the solution isotonic. To this solution was added the remaining water for injection to the desired weight, filtered through a 0.2 micron filter and packaged under aseptic conditions.
Dose and administration:
the compounds of the present invention may be formulated in a variety of oral dosage forms and carriers. Oral administration may be in the form of tablets, coated tablets, dragees, hard and soft gelatin capsules, solutions, emulsions, syrups or suspensions. The compounds of the present invention are also effective when administered by other routes of administration, including continuous (intravenous drip), topical, parenteral, intramuscular, intravenous, subcutaneous, transdermal (which may contain permeation enhancers), buccal, nasal, inhalation, and suppository administration. The preferred mode of administration is generally oral using a convenient daily dosing regimen which may be adjusted according to the extent of the disease and the patient's response to the active ingredient.
The compounds of the present invention and their pharmaceutically acceptable salts, together with one or more conventional excipients, carriers or diluents, may be formulated into pharmaceutical compositions and unit dosage forms. The pharmaceutical compositions and unit dosage forms can contain conventional ingredients in conventional proportions, with or without additional active compounds or ingredients, and the unit dosage forms can contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed. The pharmaceutical composition may be used in the form of: solids for oral use, such as tablets or filled capsules, semisolids, powders, sustained release formulations, or liquids, such as solutions, suspensions, emulsions, elixirs, or filled capsules; or suppositories for rectal or vaginal administration; or a sterile injectable solution for parenteral use. Typical formulations contain from about 5% to about 95% active compound (w/w). The term "formulation" or "dosage form" is intended to include both solid and liquid formulations of the active compound, and one skilled in the art will appreciate that the active ingredient may be present in different formulations depending on the target organ or tissue and the desired dosage and pharmacokinetic parameters.
The term "excipient" as used herein refers to a compound that is useful in preparing a pharmaceutical composition, is generally safe, non-toxic, neither biologically nor otherwise undesirable, and includes excipients that are acceptable for veterinary use as well as human pharmaceutical use. The compounds of the present invention may be administered alone, but will generally be administered with one or more suitable pharmaceutical excipients, diluents or carriers selected with regard to the intended route of administration and standard pharmaceutical practice.
"pharmaceutically acceptable" refers to materials that can be used in the preparation of pharmaceutical compositions that are generally safe, non-toxic, and neither biologically nor otherwise undesirable, and include materials that are pharmaceutically acceptable for veterinary use and for human consumption.
The "pharmaceutically acceptable salt" form of the active ingredient may also impart desirable pharmacokinetic properties to the active ingredient in that its non-salt form is not present, and may even positively influence the pharmacodynamics of the active ingredient with respect to its in vivo therapeutic activity. The phrase "pharmaceutically acceptable salt" of a compound means a salt that is pharmaceutically acceptable and possesses the desired pharmacological activity of the parent compound. Such salts include: (1) acid addition salts with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, or with organic acids; such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1, 2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo [2.2.2] oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tert-butylacetic acid, laurylsulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) a salt formed when an acidic proton present in the parent compound is replaced with a metal ion (e.g., an alkali metal ion, an alkaline earth metal ion, or an aluminum ion); or a salt formed when coordinated with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. A solid carrier can be one or more substances which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material. In powders, the carrier is usually a finely divided solid which is in admixture with the finely divided active component. In tablets, the active ingredient is usually mixed with a carrier having the required binding capacity in suitable proportions and compacted in the shape and size desired. Suitable carriers include, without limitation, magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like. Formulations in solid form may contain, in addition to the active ingredient, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.
Liquid preparations are also suitable for oral administration, and include emulsions, syrups, elixirs, aqueous solutions, and aqueous suspensions. They include solid form preparations which are intended to be converted, shortly before use, to liquid form preparations. Emulsions may be prepared in solution (e.g., in aqueous propylene glycol) or may contain emulsifying agents, for example, lecithin, sorbitan monooleate, or acacia. Aqueous solutions can be prepared by dissolving the active ingredient in water and adding suitable colorants, flavors, stabilizers, and thickening agents. Aqueous suspensions may be prepared by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending agents.
The compounds of the invention may be formulated for parenteral administration (e.g., by injection, e.g., bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, for example, solutions in aqueous polyethylene glycol. Examples of oily or nonaqueous carriers, diluents, solvents or excipients include propylene glycol, polyethylene glycol, vegetable oils (e.g., olive oil), and injectable organic esters (e.g., ethyl oleate), and may contain formulatory agents such as preservatives, wetting, emulsifying or suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in the form of a solid, sterilized by sterile packaging, or a powder, prepared by freeze-drying the solution, for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
The compounds of the invention may be prepared for topical administration to the epidermis in the form of ointments, creams or lotions, or as a transdermal patch. Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents. Formulations suitable for topical administration in the oral cavity include lozenges comprising the active ingredient in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; mouthwashes containing the active ingredient in a suitable liquid carrier.
The compounds of the present invention may also be formulated for administration as suppositories. First, a low melting wax (such as a mixture of fatty acid glycerides or cocoa butter) is melted and the active ingredient is dispersed homogeneously by, for example, stirring. The molten homogeneous mixture is then poured into a suitably sized mold, cooled and solidified.
The compounds of the invention may be prepared in a form for vaginal administration. Pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
The compounds of the invention may be formulated for nasal administration. Solutions or suspensions can be applied directly to the nasal cavity by conventional means, for example, in the form of a dropper, pipette or spray. The formulation may be in single or multiple dose form. In the case of a dropper or pipette, this may be achieved by administering an appropriate, predetermined volume of solution or suspension to the patient. In the case of spraying, this can be achieved, for example, by means of a metered atomizing spray pump.
The compounds of the invention may be prepared for aerosol administration, particularly for use in the respiratory tract, including intranasal administration. The compounds typically have a relatively small particle size, for example a particle size of 5 microns or less. The particle size may be obtained by methods known in the art, for example by micronisation. The active ingredient may be provided in a pressurised container containing a suitable propellant, such as a chlorofluorocarbon (CFC), for example dichlorodifluoromethane, trichlorofluoromethane or dichlorotetrafluoroethane or carbon dioxide or other suitable gas. The aerosol may also conveniently contain a surfactant such as lecithin. The dosage of the drug may be controlled with a metering valve. Alternatively, the active ingredient may be provided in the form of a dry powder, for example a powder mix of the compound in a suitable powder base such as lactose, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP). The powder carrier will form a gel in the nasal cavity. The powder compositions may be presented in unit dosage form, for example in capsules or cartridges of, for example, gelatin or in blister packs, from which the powder may be administered by means of an inhaler.
When desired, enteric coating materials suitable for sustained or controlled release administration of the active ingredient may be employed to prepare the formulations. For example, the compounds of the present invention may be formulated into transdermal or subcutaneous drug delivery devices. These delivery systems are advantageous when sustained release of the compound is desired and patient compliance with a treatment regimen is critical. The compounds in transdermal delivery systems are typically attached to a skin-adherent solid support. The compound of interest may also be used in combination with a penetration enhancer such as azone (1-dodecaazacycloheptan-2-one). The sustained release delivery system may be inserted subcutaneously into the subcutaneous layer by surgery or injection. Subcutaneous implants encapsulate the compound in a lipid soluble membrane such as silicone rubber or a biodegradable polymer such as polylactic acid.
In Remington: suitable formulations are described in The Science and Practice of pharmacy1995.e.w.martin, Mack Publishing Company, 19 th edition, Easton, Pennsylvania, along with pharmaceutical carriers, diluents and excipients. One skilled in the art of formulation can modify the formulation within the teachings of the specification to provide a wide variety of formulations for a particular route of administration without destabilizing the compositions of the invention or compromising their therapeutic activity.
Modifications to the compounds of the invention to provide greater solubility in water or other carriers can be readily accomplished by minor modifications (salt formation, esterification, etc.), which are well within the ordinary skill in the art. It is also well within the ordinary skill in the art to vary the route of administration and dosage regimen of a particular compound in order to control the pharmacokinetics of the compounds of the present invention in order to achieve maximum beneficial effect in the patient.
The term "therapeutically effective amount" as used herein refers to the amount required to alleviate the symptoms of a disease in an individual. The dosage can be adjusted in each particular case according to the individual requirements. The dosage may vary within wide limits depending on various factors, such as the severity of the disease being treated, the age and general health of the patient, the other drugs being used to treat the patient, the route and form of administration, and the preferences and experience of the attending medical personnel. For oral administration, a daily dose of about 0.01 to about 1000mg/kg body weight/day should be appropriate in monotherapy and/or in combination therapy. Preferred daily dosages are from about 0.1 to about 500mg/kg body weight/day, more preferably from 0.1 to about 100mg/kg body weight/day, and most preferably from 1.0 to about 10mg/kg body weight/day. Thus, for administration to a 70kg human, the dosage will range from about 7mg to 0.7 g/day. The daily dose may be administered as a single dose or in divided doses, typically 1 to 5 doses per day. In general, treatment is initiated at smaller doses than the optimal dose of the compound. The dose is then gradually increased in smaller increments until the optimum effect is achieved for the individual patient. The skilled artisan, without undue experimentation in treating the diseases described herein, will be able to determine a therapeutically effective amount of a compound of the invention for a particular disease and patient based on the knowledge, experience, and disclosure of the subject application.
The pharmaceutical preparation is preferably in unit dosage form. In such forms, the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form may be a packaged preparation, a package containing a discrete quantity of preparation such as packaged tablets, capsules and powders in vials or ampoules. The unit dosage form can also be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
Indications and treatment methods
Indications of
The compounds of the present invention, as well as isomeric forms and pharmaceutically acceptable salts thereof, are useful in the treatment and prevention of HCV infection.
The present application provides a method of treating a Hepatitis C Virus (HCV) infection comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any one of formulae I-IIII.
The present application provides a method of inhibiting HCV replication in a cell comprising administering a compound of any one of formulas I-IIII.
Combination therapy
The compounds of the present invention, as well as isomeric forms and pharmaceutically acceptable salts thereof, can be used for the treatment and prevention of HCV infection, either alone or in combination with other compounds that target viral or cellular elements involved in the HCV life cycle. The classes of compounds useful in the present invention include, without limitation, all classes of HCV antiviral agents.
For combination therapy, the types of mechanisms that can be used for active agents used in combination with the compounds of the present invention include, for example, nucleoside and non-nucleoside inhibitors of HCV polymerase, protease inhibitors, helicase inhibitors, NS4B inhibitors, and pharmaceutically active agents that functionally inhibit the Internal Ribosome Entry Site (IRES), as well as other drugs that inhibit HCV cell attachment or viral entry, HCV RNA translation, HCV RNA transcription, replication, or HCV maturation, assembly, or viral release. Specific compounds within these classes that can be used in the present invention include, without limitation, macrocyclic, heterocyclic and linear HCV protease inhibitors such as telaprevir (VX-950), boceprevir (SCH-503034), nalaprevir (naraprevir) (SCH-900518), ITMN-191(R-7227), TMC-435350(a.k.a.TMC-435), MK-7009, BI-201335, BI-1 (cilaprevir)), BMS-650032, ACH-1625, ACH-1095(HCV NS4A protease cofactor inhibitor), VX-500, VX-813, PHX-1766, PHX2054, IDX-136, IDX-316, ABT-450EP-013420 (and the like), and VBY-376; nucleoside HCV polymerase (replicase) inhibitors useful in the present invention include, without limitation, R7128, PSI-7851, IDX-184, IDX-102, R1479, UNX-08189, PSI-6130, PSI-938, and PSI-879, as well as various other nucleoside and nucleoside analogs and HCV inhibitors, including, but not limited to, those derived as 2' -C-methyl modified nucleosides (acids), 4' -aza modified nucleosides (acids), and 7' -deaza modified nucleosides (acids). Non-nucleoside HCV polymerase (replicase) inhibitors useful in the present invention include, but are not limited to, HCV-796, HCV-371, VCH-759, VCH-916, VCH-222, ANA-598, MK-3281, ABT-333, ABT-072, PF-00868554, BI-207127, GS-9190, A-837093, JKT-109, GL-59728, and GL-60667.
Furthermore, the compounds of the invention may also be combined with: cyclophilin (cyclophilin) and immunophilin (immunophilin) antagonists (such as, but not limited to, DEBIO compounds, NM-811, and cyclosporines and derivatives thereof), kinase inhibitors, heat shock protein inhibitors (such as HSP90 and HSP70), other immunomodulators, including, without limitation, interferons (-alpha, -beta, -omega, -gamma, -lambda or synthetic) such as Intron A, Roferon-A, Canferon-A300, Advaferon, Infergen, Humoferon, Sumiferon MP, Alfaferone, IFN-beta, Feron, and the like; polyethylene glycol-derivatized (PEGylated) interferon compounds, such as PEG interferon- α -2a (Pegasys), PEG interferon- α -2b (PEGIntron), PEGylated IFN- α -con1, and the like; long acting formulations and derivatives of interferon compounds such as albumin-fused interferon, Albuferon, Locteron, and the like; interferons using various types of controlled delivery systems (e.g., ITCA-638, omega-interferon delivered by DUROS subcutaneous delivery systems); compounds that stimulate interferon synthesis in cells, such as lesimett, etc.; interleukins; compounds that enhance type 1 helper T cell responses, such as SCV-07, and the like; TOLL-like receptor agonists such as CpG-10101(actilon), isotorabine, ANA773, and the like; thymosin alpha 1; ANA-245 and ANA-246; histamine dihydrochloride; propagehanium (propagermanium); tetrachlorodecaoxide; the anapril is close; IMP-321; KRN-7000; antibodies, such as civacir, XTL-6865, and prophylactic and therapeutic vaccines, such as Innovac C, HCV E1E2/MF59, and the like. In addition, any of the above methods involving the administration of an NS5A inhibitor, a type I interferon receptor agonist (e.g., IFN- α), and a type II interferon receptor agonist (e.g., IFN- γ) can be enhanced by administering an effective amount of a TNF- α antagonist. Examples of non-limiting TNF- α antagonists suitable for use in such combination therapies include ENBREL, REMICADE, and HUMIRA.
In addition, the compounds of the present invention may also be used in combination with antiprotozoal agents and other antiviral agents believed to be effective in treating HCV infection, such as, but not limited to, the prodrug nitazoxanide. Nitazoxanide can be used in the form of an agent that is combined with the compounds of the present invention and with other agents used to treat HCV infection, such as peg interferon alpha-2 a and ribavirin.
The compounds of the invention may also be used with: other forms of interferons and pegylated interferons, ribavirin or its analogs (e.g., tarabavarin, levoviron), micrornas, small interfering RNA compounds (e.g., SIRPLEX-140-N, etc.), nucleotide or nucleoside analogs, immunoglobulins, hepatoprotectors, anti-inflammatory agents, and other NS5A inhibitors. Inhibitors of other targets of the HCV life cycle include NS3 helicase inhibitors; NS4A cofactor inhibitor; antisense oligonucleotide inhibitors such as ISIS-14803, AVI-4065 and the like; vector-encoded short hairpin rna (shrna); HCV-specific ribozymes such as heptazyme, RPI, 13919, and the like; entry inhibitors such as HepeX-C, HuMax-HepC, etc.; alpha glucosidase inhibitors such as cidevir, UT-231B, etc.; KPE-02003002 and BIVN401 and an IMPDH inhibitor. Other exemplary HCV inhibitor compounds include those disclosed in the following publications, US patent Nos. 5,807,876; 6,498,178, respectively; 6,344,465 and 6,054,472; PCT patent application publication Nos. WO 97/40028; WO 98/40381; WO00/56331, WO 02/04425; WO 03/007945; WO 03/010141; WO 03/000254; WO 01/32153; WO 00/06529; WO 00/18231; WO 00/10573; WO 00/13708; WO 01/85172; WO 03/037893; WO 03/037894; WO 03/037895; WO 02/100851; WO 02/100846; WO 99/01582; WO 00/09543; WO 02/18369; WO98/17679, WO 00/056331; WO 98/22496; WO 99/07734; WO05/073216, WO05/073195 and WO 08/021927.
In addition, for example, a combination of ribavirin and interferon may be administered in the form of a multiple combination therapy for use with at least one compound of the present invention. The present invention is not limited to the above types of compounds, but contemplates combinations of known and novel compounds and bioactive agents. The combination therapy of the present invention also includes any chemically compatible combination of a compound of the present invention with other compounds of the present invention or with other compounds in addition to the compound of the present invention, so long as the combination does not abrogate the antiviral activity of the compound of the present invention or the antiviral activity of the pharmaceutical composition itself.
Combination therapy may be sequential, i.e., treatment with one active agent first, followed by treatment with a second active agent (e.g., where each treatment comprises a different compound of the invention, or where one treatment comprises a compound of the invention and the other treatment comprises one or more biologically active agents), or treatment with both active agents may be simultaneous (simultaneous). Sequential treatment may comprise a reasonable period of time after the first treatment is completed and before the second treatment is initiated. Concurrent treatment with two active agents may be carried out in the same daily dose or in separate doses. Combination therapy is not limited to two active agents and may include three or more active agents. The dosages for simultaneous or sequential combination therapy will depend on the absorption, distribution, metabolism and excretion rates of the components of the combination therapy, as well as other factors known to those skilled in the art. Dosage values will also vary depending on the severity of the condition alleviated. It will be further understood that the specific dosage regimen and schedule may be adjusted over time for any particular individual as required by the individual and at the discretion of those skilled in the art of administering or supervising the administration of the combination therapy.
The present application provides a method of treating a Hepatitis C Virus (HCV) infection comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any one of formulas I-III.
The present application provides the above method further comprising administering an immune system modulator or antiviral agent that inhibits replication of HCV, or a combination thereof.
The present application provides the above method wherein the immune system modulator is an interferon or a chemically derivatized interferon.
The present application provides the above methods, wherein the antiviral agent is selected from the group consisting of an HCV protease inhibitor, an HCV polymerase inhibitor, an HCV helicase inhibitor, an HCV primase inhibitor, an HCV fusion inhibitor, and combinations thereof.
Examples
Abbreviations
Common abbreviations include acetyl (Ac), Azobisisobutyronitrile (AIBN), atmospheric pressure (Atm), 9-borabicyclo [3.3.1]Nonane (9-BBN or BBN), 2 '-bis (diphenylphosphino) -1,1' -Binaphthyl (BINAP), tert-butoxycarbonyl (Boc), di-tert-butylpyrocarbonate or Boc anhydride (BOC2O), benzyl (Bn), butyl (Bu), chemical abstracts registration number (CASRN), benzyloxycarbonyl (CBZ or Z), Carbonyldiimidazole (CDI), 1, 4-diazabicyclo [2.2.2]Octane (DABCO), diethylaminosulfur trifluoride (DAST), dibenzylideneacetone (dba), 1, 5-diazabicyclo [4.3.0]Non-5-ene (DBN), 1, 8-diazabicyclo [5.4.0]Undec-7-ene (DBU), N ' -Dicyclohexylcarbodiimide (DCC), 1, 2-Dichloroethane (DCE), Dichloromethane (DCM), 2, 3-dichloro-5, 6-dicyano-1, 4-benzoquinone (DDQ), diethyl azodicarboxylate (DEAD), diisopropyl azodicarboxylate (DIAD), di-isobutylaluminum hydride (DIBAL or DIBAL-H), di-isopropylethylamine (DIPEA), N-Dimethylacetamide (DMA), 4-N, N-Dimethylaminopyridine (DMAP), N-Dimethylformamide (DMF), dimethyl sulfoxide (DMSO), 1' -di- (diphenylphosphino) ethane (dppe), 1' -bis- (diphenylphosphino) ferrocene (dppf), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (EDCI), 2-ethoxy-1-ethoxycarbonyl-1, 2-dihydroquinoline (EEDQ), ethyl (Et), ethyl acetate (EtOAc), ethanol (EtOH), 2-ethoxy-2H-quinoline-1-carboxylic acid Ethyl Ester (EEDQ), diethyl ether (Et)2O), ethyl isopropyl ether (EtOiPr), O- (7-azabenzotriazol-1-yl) -N, N, N 'N' -tetramethyluronium hexafluorophosphate acetic acid (HATU), acetic acid (HOAc), 1-N-hydroxybenzotriazole (HOBt), High Pressure Liquid Chromatography (HPLC), Isopropanol (IPA), isopropyl magnesium chloride (iPrMgCl), Hexamethyldisilazane (HMDS), liquid chromatography-mass spectrometry (LCMS), hexamethyldisilazaneLithium amide (LiHMDS), m-chloroperoxybenzoic acid (m-CPBA), methanol (MeOH), melting point (mp), MeSO2- (methylsulfonyl or Ms), methyl (Me), acetonitrile (MeCN), m-chloroperbenzoic acid (MCPBA), mass spectrometry (Ms), methyl tert-butyl ether (MTBE), methyltetrahydrofuran (MeTHF), N-bromosuccinimide (NBS), N-butyllithium (nBuLi), N-carboxyanhydride (NCA), N-chlorosuccinimide (NCS), N-methylmorpholine (NMM), N-methylpyrrolidone (NMP), pyridinium chlorochromate (PCC), dichloro- ((bis-diphenylphosphino) ferrocenyl) palladium (II) (Pd (dppf) Cl2) Palladium (II) acetate (Pd (OAc)2) Tris (dibenzylideneacetone) dipalladium (0) (Pd)2(dba)3) Pyridine Dichromate (PDC), phenyl (Ph), propyl (Pr), iso-propyl (i-Pr), pounds per square inch (psi), pyridine (pyr), 1,2,3,4, 5-pentaphenyl-1' - (di-tert-butylphosphino) ferrocene (Q-Phos), room temperature (ambient temperature, RT or RT), sec-butyl lithium (sBuLi), tert-butyldimethylsilyl or tert-BuMe2Si (TBDMS), tetra-n-butylammonium fluoride (TBAF), triethylamine (TEA or Et)3N), 2,6, 6-tetramethylpiperidine 1-oxyl (TEMPO), trifluoromethanesulfonate or CF3SO2- (Tf), trifluoroacetic acid (TFA), 1' -di-2, 2,6, 6-tetramethylheptane-2, 6-dione (TMHD), O-benzotriazol-1-yl-N, N, N ', N ' -tetramethylurea tetrafluorophosphate (TBTU), Thin Layer Chromatography (TLC), Tetrahydrofuran (THF), trimethylsilyl or Me3Si (TMS), p-toluenesulfonic acid monohydrate (TsOH or pTsOH), 4-Me-C6H4SO2-or tosyl (Ts) and N-urethane-N-carboxy anhydride (UNCA). When used with alkyl moieties, the conventional nomenclature including the prefixes n (n), iso (i-), secondary (sec-), tertiary (tert-), and neo (neo) have their conventional meaning. (J.Rigaudy and D.P.Klesney, Nomenclature of organic chemistry (Nomenclature in organic chemistry), IUPAC1979Pergamon Press, university of Oxford).
General conditions
The compounds of the present invention can be prepared by a variety of methods described in the illustrative synthetic reactions described in the examples section below.
The starting materials and Reagents used in preparing these compounds are generally readily available from commercial suppliers such as aldrich chemical co, or are prepared by procedures known to those skilled in the art according to procedures described in the literature, such as Fieser and Fieser's Reagents for Organic Synthesis; wiley & Sons, New York, 1991, Vol.1-15; rodd's Chemistry of Carbon Compounds, Elsevier science publishers,1989, Vol.1-5 and suppl.A; and Organic Reactions, Wiley & Sons, New York, 1991, volumes 1-40. It will be appreciated that the synthetic reaction schemes shown in the examples section are merely illustrative of some of the methods by which the compounds of the invention may be synthesized, and that various modifications may be made to these synthetic reaction schemes, as will be recognized by those skilled in the art having reference to the disclosure contained herein.
If desired, starting materials and intermediates of the synthetic reaction schemes can be isolated and purified using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography, and the like. Such materials can be characterized by conventional means including physical constants and spectral data.
Unless specifically stated to the contrary, the reactions described herein are typically carried out under an inert atmosphere, at atmospheric pressure, at a reaction temperature of from about-78 ℃ to about 150 ℃, typically from about 0 ℃ to about 125 ℃, more typically and conveniently at about room (or ambient) temperature, for example about 20 ℃.
The various substituents on the compounds of the present invention may be present in the starting compounds, added to any of the intermediates or added after the formation of the final product using known substitution methods or conversion reactions. If the substituent is reactive per se, the substituent per se may be protected by techniques known in the art. Various protecting groups are known in the art and may be used. Examples of many possible Groups can be found in "Protective Groups in organic Synthesis", Green et al John Wiley and Sons, 1999. For example, a nitro group may be introduced by nitration, and the nitro group may be converted to other groups, for example to an amino group by reduction, a halogen introduced by diazotization of the amino group and replacement of the diazo group with a halogen. The acyl group may be introduced by Friedel-Crafts acylation. The acyl group can then be converted to the corresponding alkyl group by a variety of methods including Wolff-Kishner reduction and Clemmenson reduction. The amino groups may be alkylated to form mono-and di-alkylamino groups; the mercapto and hydroxy groups may be alkylated to form the corresponding ethers. The primary alcohol may be oxidized with oxidizing agents known in the art to form a carboxylic acid or aldehyde, and the secondary alcohol may be oxidized to form a ketone. Thus, substitution or alteration reactions may be used to provide various substituents in the starting material, intermediate or end product molecule, including isolated products.
Preparation examples
Example 1
{ (2S,5S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ] indol-5-yl } -carbamic acid methyl ester
N, N' -diisopropylethylamine (3.60g,27.90mmol) was added dropwise to a heterogeneous mixture of (S) -1- (tert-butoxycarbonyl) pyrrolidine-2-carboxylic acid (2.00g,9.29mmol), 4-bromoaniline (1.60g,9.29mmol), HATU (3.53g,9.29mmol) and DMF (15ml) at room temperature. After complete addition, the reaction was stirred at room temperature for 16 hours. The reaction mixture was diluted with ethyl acetate and washed with water, 1N hydrochloric acid, saturated sodium bicarbonate solution, saturated sodium chloride solution and dried over magnesium sulfate, filtered, and concentrated to give (S) -tert-butyl 2- (4-bromophenylcarbamoyl) pyrrolidine-1-carboxylate as an off-white solid, (3.40g, 99%): ESI-LRMS m/e calculated value: C16H21BrN2O3[M+]369, found: 370[ M + H+]。
A mixture of (S) -tert-butyl 2- (4-bromophenylcarbamoyl) pyrrolidine-1-carboxylate (3.40g,9.21mmol) and 4.0M HCl/dioxane solution (15ml) in methanol (20ml) was stirred at room temperature for 4 hours. The reaction was concentrated in vacuo to give (S) -N- (4-bromophenyl) pyrrolidine-2-carboxamide hydrochloride as a pale brown solid (2.78g, 99%) ESI-LRMS m/e calculated as C11H13BrN2O HCl[M+]305.5, found: 270[ M + H+](free base).
N, N' -diisopropylethylamine (248mg,1.92mmol) was added dropwise to (2S,5S) -5- (((9H-fluoren-9-yl) methoxy) carbonylamino) -4-oxo-1, 2,4,5,6, 7-hexahydroazepino heptatrieno [3,2, 1-hi) at room temperature]Indole-2-carboxylic acid (300mg,640mmol), (S) -N- (4-bromophenyl) pyrrolidine-2-carboxamide hydrochloride (196mg,640mmol), HATU (243mg,640mmol) and DMF (15ml) in a heterogeneous mixture. After complete addition, the reaction was stirred at room temperature for 16 hours. The reaction mixture was diluted with ethyl acetate, washed with water, 1N hydrochloric acid, saturated sodium bicarbonate solution, saturated sodium chloride solution and dried over magnesium sulfate, filtered, and concentrated to give (9H-fluoren-9-yl) methyl (2S,5S) -2- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) -4-oxo-1, 2,4,5,6, 7-hexahydroazaheterocycloheptatrienoo [3,2,1-hi ] as a pale brown solid]Calculation of ESI-LRMS m/e for indol-5-yl carbamate (438mg, 95%): C39H35BrN4O5[M+]719, found value: 720[ M + H+]。
Reacting (9H-fluoren-9-yl) methyl (2S,5S) -2- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ]]A solution of indol-5-yl carbamate in 20% piperidine/DMF (10ml) was stirred at room temperature for 1 hour. The reaction was concentrated in vacuo to give (S) -1- ((2S,5S) -5-amino-4-oxo-1, 2,4,5,6, 7-hexahydroazepino [3,2, 1-hi) as a yellow solid]Indole-2-carbonyl) -N- (4-bromophenyl) pyrrolidine-2-carboxamide (170mg, 57%). The calculated value of ESI-LRMS m/e: C24H25BrN4O3[M+]497, found: 498[ M + H+]。
To ice-cold (S) -1- ((2S,5S) -5-amino-4-oxo-1, 2,4,5,6, 7-hexahydroazepino [3,2, 1-hi)]To a solution of indole-2-carbonyl) -N- (4-bromophenyl) pyrrolidine-2-carboxamide (160mg,322mmol) in DMF (10ml) was added sodium carbonate (41mg,386mmol) and methyl chloroformate (33mg,354 mmol). After complete addition, the ice bath was removed and the reaction was stirred at room temperature for 1 hour. The reaction mixture was diluted with ethyl acetate, washed with water and saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated. The resulting crude product was purified by ISCO Flash Column chromatography (TeledyneIsco RediSep Flash Column40 g; (0% to 100% ethyl acetate/hexane) to yield (2S,5S) -2- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) -4-oxo-1, 2,4,5,6, 7-hexahydroazepinoheptatrieno [3,2,1-hi ] as a white solid]Indol-5-yl carbamic acid methyl ester (64mg, 36%): c26H27BrN4O5[M+]555, found: 556[ M + H+]。
1,1' -bis (diphenylphosphino) ferrocene-palladium (II) dichloride (26mg, 36. mu. mol) was added to (2S,5S) -2- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) -4-oxo-1, 2,4,5,6, 7-hexahydroazacyclohepta [3,2,1-hi ] in one sealed tube]Indol-5-ylcarbamic acid methyl ester (200mg,360mmol), (S) -3-methyl-1-oxo-1- ((S) -2- (5- (4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -1H-imidazol-2-yl) pyrrolidin-1-yl) butan-2-ylcarbamic acid methyl ester (179mg,360mmol) and sodium bicarbonate (91mg,1.08mmol) in a mixture of 1, 2-dimethoxyethane (6ml) and water (1 ml). The reaction mixture was flushed with nitrogen, capped and heated in an oil bath (80 ℃) for 16 hours. The reaction mixture was concentrated, partitioned between 20% methanol/dichloromethane and water, and the aqueous phase was extracted with 20% methanol/dichloromethane. The combined organic phases were washed with saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated. The crude product obtained is purified by reverse phase HPLC using a 50g Polaris C18A column and is/are treated with acetonitrileWater (30% to 100%) to give { (2S,5S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] as a white solid]-3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl]-4-oxo-1, 2,4,5,6, 7-hexahydro-azepan [3,2,1-hi]Indol-5-yl } -carbamic acid methyl ester (65mg, 21%): c46H52N8O8[M+]844, found: 845[ M + H+];1H NMR(DMSO-d6)d:11.76(br.s.,1H),10.08(br.s.,1H),7.75(d,J=6.8Hz,2H),7.60-7.70(m,5H),7.43-7.53(m,2H),7.21-7.33(m,1H),6.91-7.15(m,3H),5.32(d,J=8.0Hz,1H),5.07–5.25(m,1H),4.43-4.75(m.,1H),4.02-4.14(m,1H),3.32-3.80(m,8H),2.89-3.17(m,6H),1.85-2.30(m,12H),0.79-0.96(m,6H)。
Example 2
{ (4S,7S) -4- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -6, 10-dioxo-octahydro-pyridazino [1,2-a ] [1,2] diazepin-7-yl } -carbamic acid methyl ester
To (1S,9S) -tert-butyl 9- (1, 3-dioxoisoindolin-2-yl) -6, 10-dioxooctahydro-1H-pyridazino [1,2-a ]][1,2]To a solution of diazepatriene-1-carboxylate (2.00g,4.68mmol) in ethanol (10ml) was added hydrazine (180mg,5.61 mmol). The reaction was stirred at room temperature for 3 hours. Ethanol and excess hydrazine are then concentrated in vacuo and the residue is coevaporated with ethanol to give (1S,9S) -tert-butyl 9-amino-6, 10-dioxooctahydro-1H-pyridazino [1,2-a ] as a white powder][1,2]Diazepine-1-carboxylate (1.63g, 100%): ESI-LRMSm/e calculated value: C14H23N3O4[M+]297, found: 298[ M + H ]+]。
To ice-cold (1S,9S) -tert-butyl 9-amino-6, 10-dioxooctahydro-1H-pyridazino [1,2-a ]][1,2]To a solution of diazepatriene-1-carboxylate (1.50g,5.04mmol) in DMF (15ml) was added sodium carbonate (642mg,6.05mmol) followed by methyl chloroformate (524mg,5.55 mmol). After complete addition, the ice bath was removed and the reaction was stirred at room temperature for 2 hours. The reaction mixture was diluted with ethyl acetate, washed with water, 2N hydrochloric acid solution, saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated to give (1S,9S) -tert-butyl 9- (methoxycarbonylamino) -6, 10-dioxooctahydro-1H-pyridazino [1, 2-a) as a white solid][1,2]Diazepine-1-carboxylate (1.28g, 71%). calculated as ESI-LRMS m/e: C16H25N3O6[M+]355, found: 356[ M + H ]+]。
To a solution of (1S,9S) -tert-butyl 9- (methoxycarbonylamino) -6, 10-dioxooctahydro-1H-pyridazino [1,2-a ] in dichloromethane (10ml)][1,2]To a solution of diazepatriene-1-carboxylate (1.25g,3.52mmol) was added trifluoroacetic acid (10 ml). The reaction was stirred at room temperature for 1 hour and concentrated in vacuo. Toluene (5ml) was added and the reaction was concentrated in vacuo to give (1S,9S) -9- (methoxycarbonylamino) -6, 10-dioxooctahydro-1H-pyridazino [1,2-a ] as a white solid][1,2]Diazepine-1-carboxylic acid (587mg, 68%). calculated ESI-LRMS m/e C12H17N3O6[M+]299, measured value: 300[ M + H+]。
N, N-diisopropylethylamine (254mg,1.96mmol) was added dropwise to (S) -N- (4-bromophenyl) pyrrolidine-2-carboxamide hydrochloride (200mg,654mmol), (1S,9S) -9- (methoxycarbonylamino) -6, 10-dioxooctahydro-1H-pyridazino [1,2-a ] at room temperature][1,2]Diazepatriene-1-carboxylic acid (196mg,654mmol), HATU (249mg,654mmol) and DMF (10ml) in a heterogeneous mixture. After complete addition, the reaction mixture was stirred at room temperature for 16 hours. The reaction mixture was diluted with ethyl acetate, and saturated with water, 1N hydrochloric acid, saturated sodium bicarbonate solution and saturatedThe sodium chloride solution was washed and dried over magnesium sulfate, filtered and concentrated to give (4S,7S) -4- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) -6, 10-dioxooctahydro-1H-pyridazino [1,2-a ] as a white solid][1,2]Diazepan-7-ylcarbamic acid methyl ester (150mg, 42%): calculation of ESI-LRMS m/e: C23H28BrN5O6[M+]550, found: 551[ M + H ]+]。
1,1' -bis (diphenylphosphino) ferrocene-palladium (II) dichloride (20mg, 27. mu. mol) was added to (4S,7S) -4- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) -6, 10-dioxooctahydro-1H-pyridazino [1,2-a ] in one sealed tube][1,2]Diazepan-7-ylcarbamic acid methyl ester (150mg,273mmol), (S) -3-methyl-1-oxo-1- ((S) -2- (5- (4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -1H-imidazol-2-yl) pyrrolidin-1-yl) butan-2-ylcarbamic acid methyl ester (135mg,273mmol) and sodium bicarbonate (69mg,818mmol) in a mixture of 1, 2-dimethoxyethane (6ml) and water (1 ml). The reaction mixture was flushed with nitrogen, capped and heated in an oil bath (80 ℃) for 16 hours. The reaction mixture was concentrated, partitioned between 20% methanol/dichloromethane and water, and the aqueous phase was extracted with 20% methanol/dichloromethane. The combined organic phases were washed with saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated. The resulting crude product was purified by reverse phase HPLC using 50g of Polaris C18A column eluting with acetonitrile/water (30% to 100%) to give { (4S,7S) -4- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl as a white solid]-3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl]-6, 10-dioxo-octahydro-pyridazino [1,2-a][1,2]Diazepatrien-7-yl } -carbamic acid methyl ester (58mg, 25%): c43H53N9O9[M+]839, found: 840[ M + H+];1H NMR(DMSO-d6)d:11.75(br.s.,1H),10.07(s,1H),7.75(br.s.,2H),7.59-7.66(m,6H),7.40(d,J=8.0Hz,1H),7.25(d,J=8.3Hz,1H),5.26(t,J=5.5Hz,1H),5.08(d,J=3.5Hz,1H),4.27-4.51(m,4H),4.07(t,J=8.3Hz,1H),3.60-3.85(m,2H),3.54(s,3H),3.30(s,3H),2.89-3.17(m,2H),1.53-2.35(m,18H),0.87(dd,J=18.9,6.4Hz,6H)
Example 3
{ (2S,5S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -4, 4-difluoropyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ] indol-5-yl } -carbamic acid methyl ester
N, N-diisopropylethylamine (3.17g,24.5mmol) was added dropwise to (2S,5S) -5- (methoxycarbonylamino) -4-oxo-1, 2,4,5,6, 7-hexahydroazepino [3,2,1-hi ] at room temperature]Indole-2-carboxylic acid (2.49g,8.18mmol), (S) -N- (4-bromophenyl) pyrrolidine-2-carboxamide hydrochloride (2.50g,8.18mmol), HATU (3.11g,8.18mmol) and DMF (10ml) in a heterogeneous mixture. After complete addition, the reaction mixture was stirred at room temperature for 4 hours. The reaction mixture was diluted with ethyl acetate, washed with water, 1N hydrochloric acid, saturated sodium bicarbonate solution, saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated. The resulting crude product was purified by ISCO Flash Column chromatography (Teledyne IscoRediSep Flash Column80 g; (0% to 100% methanol/dichloromethane) to give (2S,5S) -2- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) -4-oxo-1, 2,4,5,6, 7-hexahydroazepino [3,2,1-hi ] as a pale brown solid]Indol-5-yl carbamic acid methyl ester (3.40g, 75%) ESI-LRMS m/e calculated value C26H27BrN4O6[M+]555, found: 556[ M + H+]。
1,1' -bis (diphenylphosphino) ferrocene-palladium (II) dichloride (46mg, 63. mu. mol) was added to (S) -tert-butyl-4, 4-difluoro-2- (5- (4- (4,4,5, 5-tetrakis-phosphonium) -chloride in a sealed tubeMethyl-1, 3, 2-Dioxoborolan-2-yl) phenyl) -1H-imidazol-2-yl) pyrrolidine-1-carboxylate (300mg,631mmol), (2S,5S) -2- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) -4-oxo-1, 2,4,5,6, 7-hexahydroazepino [3,2,1-hi]Indol-5-ylcarbamic acid methyl ester (351mg,631mmol) and sodium bicarbonate (159mg,1.89mmol) in a mixture of 1, 2-dimethoxyethane (6ml) and water (1 ml). The reaction mixture was flushed with nitrogen, capped and heated in an oil bath (80 ℃) for 16 hours. The reaction mixture was concentrated, partitioned between 20% methanol/dichloromethane and water, and the aqueous phase was extracted with 20% methanol/dichloromethane. The combined organic phases were washed with saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated. The resulting crude product was purified by ISCO flash column chromatography (Teledyne ISCO RediSep FlashColumn40 g; (0% to 100% methanol/dichloromethane) to give (S) -tert-butyl-4, 4-difluoro-2- (5- (4' - ((S) -1- ((2S,5S) -5- (methoxycarbonylamino) -4-oxo-1, 2,4,5,6, 7-hexahydroazepinoheptane heptatrieno [3,2,1-hi ] as a white solid]Indole-2-carbonyl) pyrrolidine-2-carbamoyl) biphenyl-4-yl) -1H-imidazol-2-yl) pyrrolidine-1-carboxylate (150mg, 29%) calculated as ESI-LRMS m/e C44H47F2N7O7[M+]823, found: 824[ M + H+]。
Reacting (S) -tert-butyl-4, 4-difluoro-2- (5- (4' - ((S) -1- ((2S,5S) -5- (methoxycarbonylamino) -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ]]A mixture of indole-2-carbonyl) pyrrolidine-2-carbamoyl) biphenyl-4-yl) -1H-imidazol-2-yl) pyrrolidine-1-carboxylate (150mg,182mmol) and 4.0M HCl/dioxane solution (10ml) in methanol (15ml) was stirred at room temperature for 3 hours. The reaction was concentrated in vacuo to give (2S,5S) -2- ((S) -2- (4' - (2- ((S) -4, 4-difluoropyrrolidin-2-yl) -1H-imidazol-5-yl) biphenyl-4-ylcarbamoyl) pyrrolidine-1-carbonyl) -4-oxo-1, 2,4,5,6, 7-hexahydroazaheterocycloheptatrieno [3,2, 1-hi) as a pale yellow solid]Indol-5-yl carbamic acid methyl ester hydrochloride (135mg, 98%). calculated as ESI-LRMS m/e: C39H39F2N7O5HCl[M+]760.5, found: 724[ M + H+](free base).
N, N-diisopropylethylamine (69mg,533mmol) was added dropwise to (2S,5S) -2- ((S) -2- (4' - (2- ((S) -4, 4-difluoropyrrolidin-2-yl) -1H-imidazol-5-yl) biphenyl-4-ylcarbamoyl) pyrrolidine-1-carbonyl) -4-oxo-1, 2,4,5,6, 7-hexahydroazepino [3,2,1-hi ] at room temperature]Indol-5-ylcarbamic acid methyl ester hydrochloride (135mg,178mmol), (S) -2- (methoxycarbonylamino) -3-methylbutyric acid (31mg,178mmol), HATU (68mg,178mmol) and DMF (5ml) in a heterogeneous mixture. After complete addition, the reaction mixture was stirred at room temperature for 4 hours. The reaction mixture was diluted with ethyl acetate, washed with water, 1N hydrochloric acid, saturated sodium bicarbonate solution, saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated. The resulting crude product was purified by reverse phase HPLC using 50g Polaris C18A column eluting with acetonitrile/water (30% to 100%) to give { (2S,5S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -4, 4-difluoropyrrolidin-2-yl) as a white solid]-3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl]-4-oxo-1, 2,4,5,6, 7-hexahydro-azepan [3,2,1-hi]Indol-5-yl } -carbamic acid methyl ester (42mg, 27%). calculated as ESI-LRMS m/e C46H50F2N8O8[M+]880, found: 881[ M + H+];1H NMR(DMSO-d6)d:11.96(br.s.,1H),10.09(s,1H),7.36-7.81(m,10H),7.12(d,J=7.0Hz,1H),7.03(d,J=7.8Hz,1H),6.90-6.98(m,1H),5.23-5.35(m,1H),4.39-4.57(m,2H),4.05-4.29(m,2H),3.93(t,J=8.3Hz,1H),3.74-3.84(m,1H),3.55(s,6H),2.87-3.17(m,6H),1.82-2.28(m,10H),0.73-0.90(m,6H)
Example 4
{ (2S,5S) -2- [ (S) -2- (4' - { 5-chloro-2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ] indol-5-yl } -carbamic acid methyl ester
To solution of { (2S,5S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] in DMF (5ml)]-3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl]-4-oxo-1, 2,4,5,6, 7-hexahydro-azepan [3,2,1-hi]To a solution of indol-5-yl } -carbamic acid methyl ester (200mg,237mmol) was added N-chlorosuccinimide (38mg,284 mmol). The reaction was stirred at 50 ℃ for 16 hours. The reaction mixture was diluted with ethyl acetate, washed with water and saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated. The resulting crude product was purified by ISCO flash column chromatography (Teledyne ISCO RediSep FlashColumn40 g; (0% to 100% methanol/dichloromethane) to give { (2S,5S) -2- [ (S) -2- (4' - { 5-chloro-2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl as an off-white solid]-3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl]-4-oxo-1, 2,4,5,6, 7-hexahydro-azepan [3,2,1-hi]Indol-5-yl } -carbamic acid methyl ester (152mg, 73%): c46H51ClN8O8[M+]879, found: 880[ M + H ]+];1H NMR(DMSO-d6)d:12.55(s,1H),10.12(s,1H),7.74(s,4H),7.64(d,J=3.0Hz,4H),7.44(dd,J=16.4,8.2Hz,1H),7.30(d,J=8.3Hz,1H),7.12(d,J=7.3Hz,1H),7.03(d,J=7.5Hz,1H),6.92-6.98(m,1H),6.84(d,J=8.8Hz,1H),5.32(dd,J=10.9,2.9Hz,1H),4.95-5.04(m,1H),4.44(dd,J=8.0,4.5Hz,1H),4.00-4.15(m,2H),3.78(d,J=5.3Hz,3H),3.58-3.71(m,1H),3.54(d,J=7.3Hz,7H),2.90-3.18(m,3H),1.88-2.26(m,10H),0.81-0.90(m,6H)
Example 5
((S) -1- { (S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -pyrrolidine-1-carbonyl } -2-methyl-propyl) -carbamic acid methyl ester
1,1' -bis (diphenylphosphino) ferrocene-palladium (II) dichloride (30mg, 40. mu. mol) was added to methyl (S) -1- ((S) -2- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) pyrrolidin-1-yl) -3-methyl-1-oxobutan-2-ylcarbamate (211mg,403mmol), (S) -3-methyl-1-oxo-1- ((S) -2- (5- (4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxolan-2-yl) phenyl) -1H-imidazol-2-yl) pyrrolidin-1-yl) butane in a sealed tube Methyl 2-ylcarbamate (200mg,403mmol) and sodium bicarbonate (102mg,1.21mmol) in a mixture of 1, 2-dimethoxyethane (6ml) and water (1 ml). The reaction mixture was flushed with nitrogen, capped and heated in an oil bath (80 ℃) for 16 hours. The reaction mixture was concentrated, partitioned between 20% methanol/dichloromethane and water, and the aqueous phase was extracted with 20% methanol/dichloromethane. The combined organic phases were washed with saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated. The resulting crude product was purified by reverse phase HPLC using a 50g Polaris C18A column eluting with acetonitrile/water (30% to 100%) to give ((S) -1- { (S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl) as an off-white solid]-3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl]-pyrrolidine-1-carbonyl } -2-methyl-propyl) -carbamic acid methyl ester (83mg, 25%) calculated as ESI-LRMS m/e: c43H56N8O8[M+]812, found: 813[ M + H+];1H NMR(DMSO-d6)d:11.70(s,1H),10.30(s,1H),7.65–7.95(m,9H),7.35–7.45(m,2H),5.20–5.30(m,1H),4.55-4.65(m,1H),4.10-4.30(m,3H),3.70–3.85(m,4H),3.60(s,6H),3.30–3.50(m,2H),1.60-2.35(m,14H),0.80–0.95(m,12H)
Example 6
[ (S) -1- ((S) -2- {5- [4' - ({ (S) -1- [ (S) -1- ((R) -2-methoxycarbonylamino-2-phenyl-acetyl) -pyrrolidine-2-carbonyl ] -pyrrolidine-2-carbonyl } -amino) -biphenyl-4-yl ] -1H-imidazol-2-yl } -pyrrolidine-1-carbonyl) -2-methyl-propyl ] -carbamic acid methyl ester
Sodium carbonate (2.10g,19.80mmol) was added to a solution of sodium hydroxide (1.32g,33.10mmol) and (R) -2-amino-2-phenylacetic acid (5.00g,33.10mmol) in water (25 ml). The solution was cooled on an ice bath and methyl chloroformate (3.44g,36.40mmol) was added dropwise thereto over 15 minutes. After complete addition, the ice bath was removed and the reaction was stirred at room temperature for 3 hours. The reaction mixture was washed with ether and the aqueous phase was acidified with concentrated HCl to pH 1-2. The aqueous phase was extracted with dichloromethane, washed with water and saturated sodium chloride solution and dried over magnesium sulfate, filtered, and concentrated to give (R) -2- (methoxycarbonylamino) -2-phenylacetic acid (4.35g, 63%) as a white solid (calculated ESI-LRMS m/e: C)10H11NO4[M+]209, found: 210[ M + H+]。
N, N-diisopropylethylamine (481mg,3.72mmol) was added dropwise to a heterogeneous mixture of (S) -N- (4-bromophenyl) -1- ((S) -pyrrolidine-2-carbonyl) pyrrolidine-2-carboxamide hydrochloride (500mg,1.24mmol), (R) -2- (methoxycarbonylamino) -2-phenylacetic acid (260mg,1.24mmol), HATU (472mg,1.24mmol) and DMF (10ml) at room temperature. After complete addition, the reaction mixture was stirred at room temperature for 3 hours. The reaction mixture was diluted with ethyl acetate, washed with water, 1N hydrochloric acid, saturated sodium bicarbonate solution, saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated to give methyl (R) -2- ((S) -2- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) pyrrolidin-1-yl) -2-oxo-1-phenylethylcarbamate (640mg, 93%) as a yellow solid calculated as ESI-LRMS m/e26H29BrN4O5[M+]557, found value: 558[ M + H+]。
1,1' -bis (diphenylphosphino) ferrocene-palladium (II) dichloride (30mg, 40. mu. mol) was added to methyl (R) -2- ((S) -2- ((S) -2- (4-bromophenylcarbamoyl) pyrrolidine-1-carbonyl) pyrrolidin-1-yl) -2-oxo-1-phenylethylcarbamate (225mg,403mmol), (S) -3-methyl-1-oxo-1- ((S) -2- (5- (4- (4,4,5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) phenyl) -1H-imidazol-2-yl) pyrrolidin-1-yl) butan-2-ylcarbamate in one sealed tube Methyl ester (200mg,403mmol) and sodium bicarbonate (102mg,1.21mmol) in a mixture of 1, 2-dimethoxyethane (6ml) and water (1 ml). The reaction mixture was flushed with nitrogen, capped and heated in an oil bath (80 ℃) for 16 hours. The reaction mixture was concentrated, partitioned between 20% methanol/dichloromethane and water, and the aqueous phase was extracted with 20% methanol/dichloromethane. The combined organic phases were washed with saturated sodium chloride solution and dried over magnesium sulfate, filtered and concentrated. The resulting crude product was purified by reverse phase HPLC using a 50g Polaris C18A column eluting with acetonitrile/water (30% to 100%) to afford [ (S) -1- ((S) -2- {5- [4' - ({ (S) -1- [ (S) -1- ((R) -2-methoxycarbonylamino-2-phenyl-acetyl) -pyrrolidine-2-carbonyl) as an off-white solid]-pyrrolidine-2-carbonyl } -amino) -biphenyl-4-yl]-1H-imidazol-2-yl } -pyrrolidine-1-carbonyl) -2-methyl-propyl]Methyl carbamate (120mg, 31%): c46H54N8O8[M+]846, found: 847[ M + H+];1H NMR(DMSO-d6)d:11.70(s,1H),10.30(s,1H),7.25–8.25(m,14H),5.20–5.30(m,2H),4.55-4.65(m,1H),4.10-4.30(m,2H),3.70–3.85(m,5H),3.60(s,6H),3.30–3.50(m,2H),1.60-2.35(m,14H),0.80–0.95(m,6H)
Biological examples
Determination of HCV GT1b inhibition of replicon activity by Compounds using the replicon luciferase reporter assay
The hepatoma cell line Huh-7 was established at 2209-23 by stable transfection with a GT-1b Con1 subgenomic bicistronic replicon in Roche as previously described. A subgenomic replicon cell line was established in cured Huh7 cells from R.Bartenschlager (J Virol.2003Mar; 77(5): 3007-19). The GT-1a H77 subgenomic replicon vector pRLuc H771b75S/I (J Virol.200177:5352-59) was generated by replacing the non-structural region of the GT-1bCon1 subgenomic replicon with the non-structural region in strain H77 (except for the first 75 amino acids of the NS3 protein of GT-1b Con 1). The GT-1a pRLuc H771b75S/I subgenomic replicon cell line was established in cured Huh7 cells from R.Bartenschlager (J Virol.2003Mar; 77(5): 3007-19).
All subgenomic replicon cell lines were grown in Dulbecco's modified eagle's Medium (DMEM-Glutamax)TM-I; invitrogen Cat # 10569-010). The medium was supplemented with 10% fetal bovine serum (Invitrogen Cat #10082-147), 1% penicillin/streptomycin (Mediatech Cat #30-002-CI) and 500. mu.g/ml G418(Mediatech Cat # 30-234-CI). Cells were maintained in humidified 5% CO at 37 ℃2In an atmosphere.
2209-23 cells were plated into 96-well plates (Becton Dickinson, Cat #353296) at a cell density of 5000 cells per well. Cells were plated onto 90. mu.l Dulbecco's modified eagle's medium (DMEM-Glutamax)TM-I), (Invitrogen Cat #10569-010), the medium was supplemented with 5% fetal bovine serum (Invitrogen Cat #10082-147), 1% penicillin/streptomycin (Mediatech Cat # 30-002-CI). pRluc H771b75S/I cells plated at 3000 cells/well into 96-well plates in 90. mu.l final volume of DMEM-Glutamax containing 5% FBS and 1% penicillin/streptomycinTM-I. These cells were allowed to stand at 37 ℃ and 5% CO2Let down for 24 hours, then add compound. Compound (or medium as control) was added 24 hours after plating in a volume of 10 μ l as a 3-fold dilution, with a final DMSO concentration of 1%. Renilla luciferase reporter signal was read 72 hours after compound addition using the Renilla luciferase assay system (Promega, cat # E2820). EC50 values were defined as the values under which Renilla fluorescence was observed compared to control samples in the absence of compoundCompound concentrations with 50% reduction in the level of the protease reporter gene as determined by non-linear fit of compound dose-response data. EC50 is approximate if the maximum percent inhibition is below 90% and above 70%.
Cytotoxicity of compounds tested using the HCV GT1b replicon cell line assay
WST12209-23 cells were plated at a cell density of 5000 cells per well into clear flat bottom 96-well plates (Becton Dickinson, Cat #353075) for cell viability studies. Cell viability was determined using the WST-1 cell proliferation assay (Roche diagnostic, Cat # 11644807001). The format of the test plate was set to be the same as in the replicon test. After 3 days incubation with compound, 10. mu.l WST-1 reagent was added to each well according to the manufacturer's instructions and incubated at 37 ℃ and 5% CO2The cells were incubated for 2 hours. The absorption reading at 450nm (reference filter at 650nm) was determined with an MRX reduction microtiter plate reader (Lab System). CC (challenge collapsar)50Values are defined as the concentration of compound required to reduce cell viability by 50% compared to an untreated control in the absence of compound, as determined by non-linear fitting of compound dose-response data. Representative experimental data can be found in table II below:
table II.
The invention has been described in detail by way of illustration and example for purposes of clarity and understanding. It will be apparent to those skilled in the art that changes and modifications may be made without departing from the scope of the appended claims. Accordingly, it is to be understood that the above description is intended to be illustrative, and not restrictive. The scope of the invention should, therefore, be determined not with reference to the above description, but instead should be determined with reference to the appended claims, along with the full scope of equivalents to which such claims are entitled.
All patents, patent applications, and publications cited in this application are incorporated herein by reference in their entirety for all purposes to the same extent as if each individual patent, patent application, or publication were specifically and individually indicated to be incorporated herein by reference.
Claims (15)
1. A compound of formula I-III or a pharmaceutically acceptable salt thereof
Wherein:
a is
In the formulae I and IIn I, each R1Independently selected from H or C1-6Alkyl, in the formula III, each R1Independently selected from H, C1-6Alkyl or phenyl;
each R2Independently selected from H or C1-6An alkyl group;
each R3Is C (═ O) OR4;
R4Is C1-6An alkyl group;
each X is independently selected from H and Cl; and is
Each Y is1And Y2Independently selected from H or F.
2. The compound of claim 1 wherein X is H, each R2Is H, and each R3Is C (═ O) OCH3。
3. The compound of claim 2, wherein R1Is isopropyl.
4. The compound of claim 3, wherein Y1Is H.
5. The compound of claim 4, wherein Y is2Is H.
6. The compound of claim 3, wherein Y1Is F.
7. The compound of claim 6, wherein Y2Is H.
8. The compound of claim 6, wherein Y2Is F.
9. A compound selected from the group consisting of:
{ (2S,5S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ] indol-5-yl } -carbamic acid methyl ester;
{ (4S,7S) -4- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -6, 10-dioxo-octahydro-pyridazino [1,2-a ] [1,2] diazepin-7-yl } -carbamic acid methyl ester;
{ (2S,5S) -2- [ (S) -2- (4' - {2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -4, 4-difluoropyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ] indol-5-yl } -carbamic acid methyl ester; and
{ (2S,5S) -2- [ (S) -2- (4' - { 5-chloro-2- [ (S) -1- ((S) -2-methoxycarbonylamino-3-methyl-butyryl) -pyrrolidin-2-yl ] -3H-imidazol-4-yl } -biphenyl-4-ylcarbamoyl) -pyrrolidine-1-carbonyl ] -4-oxo-1, 2,4,5,6, 7-hexahydro-azepino [3,2,1-hi ] indol-5-yl } -carbamic acid methyl ester.
10. Use of a compound according to any one of claims 1-9 in the manufacture of a medicament for the treatment of Hepatitis C Virus (HCV) infection.
11. The use of claim 10, said medicament being co-administered with an immune system modulator or an antiviral agent that inhibits replication of HCV, or a combination thereof.
12. The use of claim 11, wherein the immune system modulator is an interferon or a chemically derivatized interferon.
13. The use of claim 11, wherein said antiviral agent is selected from the group consisting of HCV protease inhibitors, HCV polymerase inhibitors, HCV helicase inhibitors, HCV primase inhibitors, HCV fusion inhibitors, and combinations thereof.
14. Use of a compound according to any one of claims 1-9 in the manufacture of a medicament for inhibiting HCV replication in a cell.
15. A composition comprising a compound according to any one of claims 1 to 9 and a pharmaceutically acceptable carrier.
Applications Claiming Priority (3)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US201161576641P | 2011-12-16 | 2011-12-16 | |
| US61/576,641 | 2011-12-16 | ||
| PCT/EP2012/075322 WO2013087743A1 (en) | 2011-12-16 | 2012-12-13 | Inhibitors of hcv ns5a |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| HK1200847A1 HK1200847A1 (en) | 2015-08-14 |
| HK1200847B true HK1200847B (en) | 2018-03-09 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| CN103987723B (en) | Inhibitors of HCV NS5A | |
| JP5808496B2 (en) | Antiviral compounds | |
| CN104185624B (en) | Antiviral compound | |
| US20120328565A1 (en) | Antiviral compounds | |
| WO2012123298A1 (en) | Antiviral compounds | |
| HK1216639A1 (en) | N-heteroaryl substituted aniline derivatives as hcv-antivirals | |
| JP2016510050A (en) | Antiviral compounds | |
| CN105026386B (en) | Antiviral compound | |
| HK1200847B (en) | Inhibitors of hcv ns5a | |
| HK1204618B (en) | Antiviral compounds |