OA17337A - Antiviral compounds. - Google Patents

Antiviral compounds. Download PDF

Info

Publication number
OA17337A
OA17337A OA1201500241 OA17337A OA 17337 A OA17337 A OA 17337A OA 1201500241 OA1201500241 OA 1201500241 OA 17337 A OA17337 A OA 17337A
Authority
OA
OAPI
Prior art keywords
compound
methyl
pyran
imidazol
tert
Prior art date
Application number
OA1201500241
Inventor
Elizabeth M. Bacon
Jeromy J. Cottell
John 0. LINK
Teresa Alejandra Trejo Martin
Original Assignee
Gilead Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Publication of OA17337A publication Critical patent/OA17337A/en

Links

Abstract

The disclosure is related to anti-viral compounds, compositions containing such compounds, and therapeutic methods that include the administration of such compounds, as well as to processes and intermediates useful for preparing such compounds.

Description

The présent disclosure provides compounds for use in pharmaceutical compositions and methods for treating hepatitis C (HCV). In particular, provided herein are compounds having a polycyclïc core and at least one 2,6-dimethyltetrahydro-2H-pyran-4-yl, 4methyltetrahydro-2H-pyran-4-yl, or tetrahydro-2H-pyran-3-y! capping group, which compounds exhibit bénéficiai properties, such as, for example, enhanced bioavailability and/or enhanced activity against certain HCV génotypes, including but not limited to, known résistant mutations thereof (see, e.g., Tables 1,2A and 2B).
In one embodiment the disclosure provides a compound of formula (I):
E’*-Vla -C^oy-P1* -W1* -P,b-C(=O)-Vlb-Elb (I) wherein:
Wis
and W1* is optionally substituted with one or more halo, alkyl, haloalkyl, optionally substituted aryl, optionally substituted heterocycle, or cyano;
Y5 is -O-CH2-, -CH2-O-, -O-C(=O>, or -C(=O)-O-;
X5 is -CH2-CH2-, or -CH=CH-;
P1* and Plb are each independently:
.AMJ MW
MW
V1* and Vlb are each independently:
provided that at least one of V1* and V,b is
Eu and Elb are each independently -N(H)(alkoxycarbonyl),
-N(H)(cycloalkylcarbonyl), or -N(HXcycloalkyloxycarbonyl); or E^-V1* taken together are R9*; or Elb-V’b taken together are R9b; and
R9 and R9b are each independently:
or a pharmaceutically acceptable sait or prodrug thereof.
The disclosure also provides isotopically enriched compounds that are compounds of the disclosure that comprise an enriched isotope at one or more positions in the compound.
The présent disclosure also provides a pharmaceutical composition comprising a compound of the disclosure or a pharmaceutically acceptable sait or prodrug thereof and at least one pharmaceutically acceptable carrier.
The présent disclosure also provides a pharmaceutical composition for use in treating hepatitis C (HCV). In one embodiment the composition comprises at least one additional therapeutic agent for treating HCV. In one embodiment, the therapeutic agent is selected from ribavirin, an NS3 protease inhibitor, a nucleoside or nucléotide inhibitor of HCV NS5B polymerase, an alpha-glucosidase 1 inhibitor, a hepatoprotectant, a non-nucleosîde inhibitor of HCV polymerase, or combinations thereof. In one embodiment, the composition further comprises a nucleoside or nucléotide inhibitor of HCV NS5B polymerase. In one embodiment, the nucleoside or nucléotide inhibitor ofHCV NS5B polymerase is selected from ribavirin, viramidine, levovirin, a L-nucleoside, or isatoribine.
In one embodiment, provided is a pharmaceutical composition comprising a compound as described herein and at least one nucleoside or nucléotide inhibitor of HCV NS5B polymerase, and at least one pharmaceutically acceptable carrier. In one embodiment, the composition further comprises an interferon, a pegylated interferon, ribavirin or combinations thereof. In one embodiment, the nucleoside or nucléotide inhibitor of HCV NS5B polymerase is sofosbuvir. In one embodiment, provided is a pharmaceutical composition comprising a compound as described herein and at least one NS3 protease inhibitor, and at least one pharmaceutically acceptable carrier. In one embodiment, the composition further comprises sofosbuvir.
The présent disclosure also provides a pharmaceutical composition further comprising an interferon or pegylated interferon.
The présent disclosure also provides a pharmaceutical composition further comprising a nucleoside analog.
The présent disclosure also provides for a pharmaceutical composition wherein said nucleoside analogue is selected from ribavirin, viramidine, levovirin, an L-nucleoside, and isatoribine and said interferon is α-interferon or pegylated a-interferon.
The présent disclosure also provides for a method of treating hepatitis C, said method comprising administering to a human patient a pharmaceutical composition which comprises a therapeutically effective amount of a compound ofthe disclosure.
The présent disclosure also provides a method of inhibiting HCV, comprising administering to a mammat afïlicted with a condition associated with HCV activity, an amount of a compound ofthe disclosure, effective to inhibit HCV.
The présent disclosure also provides a compound of the disclosure for use in medical therapy (e.g. for use in inhibiting HCV activity or treating a condition associated with HCV activity), as well as the use of a compound of the disclosure for the manufacture of a médicament useful for inhibiting HCV or the treatment of a condition associated with HCV activity in a mammal.
The présent disclosure also provides synthetic processes and noveî intermediates disclosed herein which are useful for preparing compounds of the disclosure. Some of the compounds of the disclosure are useful to préparé other compounds of the disclosure. In another aspect the disclosure provides a compound of the disclosure, or a pharmaceutically acceptable sait or prodrug thereof, for use in the prophylactic or therapeutic treatment of hepatitis C or a hepatitis C associated disorder.
In another aspect the disclosure provides a method of inhibiting HCV activity in a sample comprising treating the sample with a compound of the disclosure.
Compounds of formula (I) hâve been found to possess useful activity against several HCV génotypes. Additionally certain compounds of formula (I) exhibit significant potency against résistant variants in, e.g., GT1.
DETAILED DESCRIPTION
Reference will now be made in detail to certain embodiments of the disclosure, examples ofwhich are illustrated in the accompanying structures and formulas. While the disclosure will be described in conjunction with the enumerated embodiments, it will be understood that they are not intended to limit the disclosure to those embodiments. On the contrary, the disclosure is intended to cover ail alternatives, modifications, and équivalents, which may be included within the scope of the présent disclosure as defîned by the embodiments.
Compounds
The “P” groups (e.g., P1* and Plb) defîned for formula (I) herein hâve one bond to a -C(=O)- of formula (I) and one bond to a Wu group. It is to be understood that a nitrogen of the P group is connected to the -C(=O)- group of formula (I) and that a carbon of the P group is connected to the W,a group.
In the W’a group, a Y3 group is présent. When that Y5 group is defîned as -O-CH2-, or -CH2-O- group, those Y5 groups hâve a directionality. The Y5 group is connected to the W1* group în the same left to right directionality that each is drawn. So for exemple, when Y’ is -O-CH2-, the directly following structure is intended:
For example, when Y5 is -CH2-O-, the directly following structure is intended:
In the structure I, the W11 group has a lefl-to-right directionality as depicted in I and W1* as they are drawn.
Ε'·.ν'« -C^O^P1* -W11 -P,b-C(=O)-Vlb-EIb (I) wherein:
W1· is
For example, the P1* group is connected to the imidazole group of W and the Plb group is connected to the pentacyclic ring system of W1*.
“Alkyi” is C1-C18 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms. Examples are methyl (Me, -CH3), ethyl (Et, -CH2CH3), 1-propyl (n-Pr, npropyl, -CH2CH2CH3), 2-propyl (i-Pr, i-propyl, -CH(CH3)2), 1-butyl (n-Bu, n-butyl, -CH2CH2CH2CH3), 2-methyl-l-propyl (i-Bu, i-butyl, -CH2CH(CH3)2), 2-butyl (s-Bu, sbutyl, -CH(CH3)CH2CH3), 2-methyl-2-propyI (t-Bu, t-butyl, -C(CH3)3), 1-pentyl (n-pentyl, -CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-penty! (-CH(CH2CH3)2), 2methyl-2-butyl (-C(CH3)2CH2CH3), 3-methyl-2-butyl (-CH(CH3)CH(CH3)2), 3-methyl-lbutyl (-CH2CH2CH(CH3)2), 2-methyl-1-butyl (-CH2CH(CH3)CH2CH3), 1-hexyl (-CH2CH2CH2CH2CH2CH3), 2-hexyl (-CH(CH3)CH2CH2CH2CH3), 3-hexyl (-CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (-C(CH3)2CH2CH2CH3), 3-methyl-2 pentyl (-CH(CH3)CH(CH3)CH2CH3), 4-methyl-2-pentyl (-CH(CH3)CH2CH(CH3)2), 3methyl-3-pentyl (-C(CH3)(CH2CH3)2), 2-methyl-3-pentyl (-CH(CH2CH3)CH(CH3)2), 2,3dimethyl-2-butyl (-C(CH3)2CH(CH3)2), 3,3-dimethyl-2-butyl (-CH(CH3)C(CH3)3, and cyclopropylmethyl “Alkenyl” is C2-C18 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, Le. a carbon-carbon, sp2 double bond. Exemples include, but are not limited to, ethylene or vinyl (-CH=CH2), allyl (-CH2CH=CH2), cyclopentenyl (-C5H7), and 5-hexenyl (-CH2 CH2CH2CH2CH=CH2) “Alkynyl” is C2-C18 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one site of unsaturation, Le. a carbon-carbon, sp triple bond. Examples include, but are not limited to, acetylenic (-C=CH) and propargyl (-CH2OCH).
“Alkylene” refers to a saturated, branched or straight chain or cyclic hydrocarbon radical of l-l 8 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. Typical alkylene radical s include, but are not limited to, methylene (-CH^) 1,2-ethyl (-CH2CH2-), 1,3propyl (-CH2CH2CH2-), 1,4-butyl (-CH2CH2CH2CH2-), and the like.
“Alkenylene” refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkene. Typical alkenylene radicals include, but are not limited to, 1,2-ethylene (-CH=CH-).
“Alkynylene” refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkyne. Typical alkynylene radicals include, but are not limited to, acetylene (-C=C-), propargyl (-CH2C=C-), and 4-pentynyl (-CH2CH2CH2C=CH).
The term alkoxy or “alkyloxy,” as used herein, refers to an alkyl group attached to the parent molecular moiety through an oxygen atom.
The term alkoxycarbonyt, as used herein, refers to an alkoxy group attached to the parent molecular moiety through a carbonyl group.
The term cycloalkyl, as used herein, refers to a saturated monocyclic, hydrocarbon ring System having three to seven carbon atoms and zéro heteroatoms. Représentative examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclopentyl, and cyclohexyl. The cycloalkyl groups ofthe présent disclosure are optionally substituted with one, two, three, four, or five substituents independently selected from alkoxy, alkyl, aryl, cyano, halo, haloalkoxy, haloalkyl, heterocyclyl, hydroxy, hydroxyalkyl, nitro, and -NR*Ry wherein the aryl and the heterocyclyl are further optionally substituted with one, two, or three substituents independently selected from alkoxy, alkyl, cyano, halo, haloalkoxy, haloalkyl, hydroxy, and nitro.
The term “cycloalkylcarbonyl,” as used herein, refers to a cycloalkyl group attached to the parent molecular moiety through a carbonyl group.
The term cycloalkyloxy, as used herein, refers to a cycloalkyl group attached to the parent molecular moiety through an oxygen atom.
The term “cycloalkyloxycarbonyl,” as used herein, refers to a cycloalkyloxy group attached to the parent molecular moiety through a carbonyl group.
“Aryl” means a monovalent aromatic hydrocarbon radical of 6-20 carbon atoms derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring System. Typical aryl groups include, but are not limited to, radicals derived from benzene, substituted benzene, naphthalene, anthracene, biphenyl, and the like.
“Arylalkyl” refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl radical. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-l-yl, naphthylmethyl, 2-naphthy!ethan-l-yl, naphthobenzyl, 2-naphthophenyIethan-l-yl and the like. The arylalkyl group comprises 6 to 20 carbon atoms, e.g., the alkyl moiety, including alkanyl, alkenyl or alkynyl groups, ofthe arylalkyl group is 1 to 6 carbon atoms and the aryl moiety is 5 to 14 carbon atoms.
“Substituted alkyl”, “substituted aryl”, and “substituted arylalkyl” mean alkyl, aryl, and arylalkyl respectively, in which one or more hydrogen atoms are each independently replaced with a non-hydrogen substituent. Typical substituents include, but are not limited to: halo (e.g. F, Cl, Br, I), -R, -OR, -SR, -NRi, -CFj, -CCb, -OCFj, -CN, -NO2, -N(R)C(=O)R, -C(=O)R, -OC(=O)R, -C(O)OR, -C(=O)NRR, -S(=O)R, -S(=O)iOR, -S(=O)?R,
-OS(=0)20R, -S(=O)2NRR, and each R is independently -H, alkyl, aryl, arylalkyl, or heterocycle. Alkylene, alkenylene, and alkynylene groups may also be similarly substîtuted. The term “optionally substîtuted” in référencé to a particular moiety of the compound of formula (I), (e.g., an optionally substîtuted aryl group) refers to a moiety having 0,1,2, or more substitueras.
The symbol “----“ in a ring structure means that a bond is a single or double bond. In
“Haloalkyl” as used herein includes an alkyl group substîtuted with one or more halogens (e.g. F, Cl, Br, or I). Représentative examples of haloalkyl include trifluoromethyl, 2,2,2-trifluoroethyl, and 2,2,2-trifluoro-l-(trifluoromethyl)ethyl.
“Heterocycle” or “heterocyclyl” as used herein includes by way of example and not limitation these heterocycles described in Paquette, Léo A.; Principles of Modem Heterocyclic Chemistry (W.A. Benjamin, New York, 1968), particularly Chapters 1,3,4, 6, 7, and 9; The Chemistry of Heterocyclic Compounds, A Sériés of Monographs” (John Wiley & Sons, New York, 1950 to présent), in particular Volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960)82:5566. In one spécifie embodiment, “heterocycle” includes a “carbocycle” as defined herein, wherein one or more (e.g. 1, 2, 3, or 4) carbon atoms hâve been replaced with a heteroatom (e.g. O, N, or S). The term heterocycle also includes “heteroaryl” which is a heterocycle wherein at least one heterocyclic rings is aromatic.
Examples of heterocycles include by way of example and not limitation pyridyl, dihydropyridyl, tetrahydropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofiiranyl, thianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, 2-pyrrolidonyl, pyrrolinyl, tetrahydrofùranyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, octahydroisoquinolinyl, azocinyl, triazinyl, 6H-l,2,5-thiadiazinyl, 2H,6H-l,5,2-dithîazinyl, thienyl, thianthrenyl, pyranyl, isobenzofuranyt, chromenyl, xanthenyl, phenoxathinyl, 2H-pyrrolyI, isothiazolyl, isoxazolyl, pyrazinyl, pyridazinyl, indolizinyl, isoindolyl, 3H-indoIyl, lH-indazolyl, purinyl, 4H-quinolizinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, quinazolinyl, cinnolinyl, pteridinyl, 4H-carbazolyl, carbazolyl, β-carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, furazanyl, phenoxazinyl, isochromanyl, chromanyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperazinyl, indolinyl, isoindolînyl, quinuclidinyl, morpholinyl, oxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, isatinoyl, and bis-tetrahydrofùranyl:
</
By way of example and not limitation, carbon bonded heterocycles are bonded at position 2,3,4, 5, or 6 of a pyridine, position 3,4,5, or 6 of a pyridazine, position 2,4, 5, or 6 of a pyrimidine, position 2,3,5, or 6 of a pyrazine, position 2, 3,4, or 5 of a fiiran, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole, position 2,4, or 5 of an oxazole, imidazole orthiazole, position 3,4, or 5 of an isoxazole, pyrazole, or isothiazole, position 2 or 3 of an aziridine, position 2,3, or 4 of an azetidine, position 2,3,4, 5,6,7, or 8 of a quinoline or position 1, 3,4, 5, 6, 7, or 8 of an isoquinoline. Still more typically, carbon bonded heterocycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazînyl, 2-pyrimidinyl, 4-pyrimidiny], 5pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4thiazolyl, or 5-thiazoIyL
By way of example and not limitation, nitrogen bonded heterocycles are bonded at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrrolinc, imidazole, imidazolidine, 2-imidazolîne, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3pyrazoline, piperidine, piperazine, indole, indoline, lH-indazole, position 2 of a isoindole, or isoindoline, position 4 ofa moTpholine, and position 9 ofa carbazole, or β-carboline. Still more typically, nitrogen bonded heterocycles include 1-aziridyl, 1-azetedyl, l-pyrrolyl, 1imidazolyl, 1-pyrazolyl, and 1-piperidinyl.
“Carbocycle” refers to a saturated, unsaturated or aromatic ring havîng up to about 25 carbon atoms. Typically, a carbocycle has about 3 to 7 carbon atoms as a monocycle, about 7 to 12 carbon atoms as a bicycle, and up to about 25 carbon atoms as a polycycle. Monocyclic carbocycles typically hâve 3 to 6 ring atoms, still more typically 5 or 6 ring atoms. Bicyclic carbocycles typically hâve 7 to 12 ring atoms, e.g., arranged as a bicyclo [4,5], [5,5], [5,6] or [6,6] system, or 9 or 10 ring atoms arranged as a bicyclo [5,6] or [6,6] System. The term carbocycle includes “cycloalkyl” which is a saturated or unsaturated carbocycle. Examples of monocyclic carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-l-enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-l-enyl, l-cyclohex-2-enyl, 1cydohex-3-enyl, phenyl, spiry! and naphthyl.
The term “amino, as used herein, refers to -NH2.
The term “chiral” refers to molécules which hâve the property of nonsuperimposability of the minor image partner, while the term “achiral” refers to molécules which are superimposable on their mirror image partner.
The term “stereoisomers” refers to compounds which hâve identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
“Diastereomer” refers to a stereoisomer with two or more cent ers of chiral ity and whose molécules are not mirror images of one another. Diastereomers hâve different physicat properties, e.g., melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers may separate under high resolution analytical procedures such as, for example, electrophoresis and chromatography.
“Enantiomers” refer to two stereoisomers of a compound which are nonsuperimposable mirror images of one another.
The term “treatment” or “treating,” to the extent it relates to a disease or condition includes preventing the disease or condition from occurring, inhibiting the disease or condition, eliminating the disease or condition, and/or relieving one or more symptoms of the disease or condition.
Stereochemical définitions and conventions used herein generally follow S. P. Parker, Ed, McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds (1994) John Wiley & Sons, Inc., New York Many organic compounds exist in optically active forms, Le., they hâve the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the préfixés (D and L) or (R and S) are used to dénoté the absolute configuration of the molécule about its chiral center(s). The préfixés d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A spécifie stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms “racemic mixture and “racemate” refer to an equimolar mixture oftwo enantiomeric species, devoid ofoptical activity. The disclosure includes ail stereoisomers of the compounds described herein.
Prodrugs
The term “prodrug” as used herein refers to any compound that when administered to a biological System generates a compound of the disclosure that inhibits HCV activity (“the active inhibitory compound). The compound may be formed from the prodrug as a resuit of: (i) spontaneous chemical reaction(s), (ii) enzyme catalyzed chemical reaction(s), (iii) photolysis, and/or (iv) metabolic chemical reaction(s).
“Prodrug moiety” refers to a labile functiona! group which séparâtes from the active inhibitory compound during metabolism, systemically, inside a cell, by hydrolysis, enzymatic cleavage, or by some other process (Bundgaard, Hans, “Design and Application of Prodrugs” in A Textbook of Drug Design and Development (1991), P. Krogsgaard-Larsen and H. Bundgaard, Eds. Harwood Academie Publishers, pp. 113-191). Enzymes which are capable of an enzymatic activation mechanism with the prodrug compounds ofthe disclosure include, but are not limited to, amidases, esterases, microbial enzymes, phospholipases, cholinesterases, and phosphases. Prodrug moieties can serve to enhance solubility, absorption and lipophilicity to optimîze drug delivery, bîoavailability and efficacy. A prodrug moiety may include an active métabolite or drug itself.
Exemplary prodrug moieties include the hydrolytically sensitive or labile acyloxymethyl esters -CH2OC(=O)R and acyloxymethyl carbonates ~CH2OC(=O)OR” where R is Ci-Ce alkyl, Ci-Ce substituted alkyl, Cô-Cîû aryl or Ce-Cîo substituted aryl. The acyloxyalkyl ester was first used as a prodrug strategy for carboxylic acids and then applied to phosphates and phosphonates by Farquhar et al. (1983)/ Pharm. Sci. 72: 324; also US Patent Nos. 4816570,4968788, 5663159 and 5792756. Subsequently, the acyloxyalkyl ester was used to deliver phosphonic acids across cell membranes and to enhance oral bîoavailability. A close variant of the acyloxyalkyl ester, the alkoxycarbonyloxyalkyl ester (carbonate), may also enhance oral bîoavailability as a prodrug moiety in the compounds ofthe combinations ofthe disclosure. An exemplary acyloxymethyl ester is pivaloyloxymethoxy, (POM) -CH2OC(=O)C(CHj)3. An exemplary acyloxymethyl carbonate prodnig moiety is pivaloyloxymethylcarbonate (POC) -CH2OC(=O)OC(CH3)3.
Protecting Groups
In the context of the présent disclosure, protecting groups include prodrug moîeties and chemical protecting groups.
“Protecting group” refers to a moiety of a compound that masks or alters the properties of a fùnctional group or the properties of the compound as a whole. Chemical protecting groups and strategies for protection/deprotection are well known in the art. See e.g., Protective Groups in Organic Chemistry, Theodora W. Greene, John Wiley & Sons, Inc., New York, 1991. Protecting groups are often utilized to mask the reactivity of certain fùnctional groups, to assist in the efficiency of desired chemical reactions, e.g., making and breaking chemical bonds in an ordered and planned fashion. Protection of fùnctional groups of a compound alters other physical properties besides the reactivity of the protected fùnctional group, such as, for example, the polarity, Jipophilicity (hydrophobicity), and other properties which can be measured by common analytical tools. Chemically protected intermediates may themselves be biologically active or inactive.
Protected compounds may also exhibit altered, and in some cases, optimized properties in vitro and in vivo, such as, for example, passage through cellular membranes and résistance to enzymatic dégradation or séquestration. In this rôle, protected compounds with intended therapeutic efïects may be referred to as prodrugs. Another fonction ofa protecting group is to convert the parental drug into a prodrug, whereby the parental drug is released upon conversion of the prodrug in vivo. Because active prodrugs may be absorbed more effectively than the parental drug, prodrugs may possess greater potency in vivo than the parental drug. Protecting groups are removed either in vitro, in the instance of chemical intermediates, or in vivo, in the case of prodrugs. With chemical intermediates, it is not particularly important that the resultîng products after deprotection, e.g., alcohols, be physiologically acceptable, although in general it is more désirable if the products are pharmacologically innocuous.
Protecting groups are available, commonly known and used, and are optionally used to prevent side reactions with the protected group during synthetic procedures, i.e. routes or methods to préparé the compounds of the disclosure. For the most part the decision as to which groups to protect, when to do so, and the nature of the chemical protecting group “PG” will be dépendent upon the chemistry of the reaction to be protected against (e.g., acidic, basic, oxidative, reductive or other conditions) and the intended direction of the synthesis. PGs do not need to be, and generally are not, the same if the compound is substituted with multiple PG. In general, PG will be used to protect functional groups such as, for example, carboxyl, hydroxyl, thio, or amino groups and to thus prevent side reactions or to otherwise facilitate the synthetic efïiciency. The order of deprotection to yield free deprotected groups is dépendent upon the intended direction of the synthesis and the reaction conditions to be encountered, and may occur in any order as determined by the artisan.
Various functional groups of the compounds of the disclosure may be protected. For example, protecting groups for -OH groups (whether hydroxyl, carboxylic acid, phosphonic acid, or other fonctions) include “ether- or ester-forming groups”. Ether- or ester-forming groups are capable of fonctioning as chemical protecting groups in the synthetic schemes set forth herein. However, some hydroxyl and thio protecting groups are neither ether- nor esterforming groups, as will be understood by those skilled in the art, and are included with amides, discussed below.
A very large number of hydroxyl protecting groups and amide-forming groups and corresponding chemical cleavage reactions are described in Protectlve Groups in Organic Synthesis, Theodora W. Greene (John Wiley & Sons, Inc., New York, 1991, ISBN 0-47162301-6) (“Greene”). See also Kocienski, Philip J.; Protecting Groups (Georg Thieme Verlag Stuttgart, New York, 1994), which is incorporated by reference in its entirety herein. In particular Chapter 1, Protecting Groups: An Overview, pages 1-20, Chapter 2, Hydroxyl Protecting Groups, pages 21-94, Chapter 3, Diol Protecting Groups, pages 95-117, Chapter 4, Carboxyl Protecting Groups, pages 118-154, Chapter 5, Carbonyl Protecting Groups, pages 155-184. For protecting groups for carboxylic acid, phosphonic acid, phosphonate, sulfonic acid and other protecting groups for acids see Greene as set forth below.
Stereoisomers
The compounds of the disclosure may hâve chiral centers, e.g., chiral carbon or phosphores atoms. The compounds of the disclosure thus include ail stereoisomers, including enantiomers, diastereomers, and atropîsomers. In addition, the compounds of the disclosure include enriched or resolved optical isomers at any or ail asymmetrîc, chiral atoms. In other words, the chiral centers apparent from the depictions are provided as the nonracemic or racemic mixtures. Both racemic and diastereomeric mixtures, as well as the individual optical isomers isolated or synthesîzed, substantially free of their enantiomeric or diastereomeric partners, are ail within the scope of the disclosure. The racemic mixtures are separated into their individual, substantially optically pure isomers through well-known techniques such as, for example, the séparation of diastereomeric salts formed with optically active adjuncts, e.g., acids or bases followed by conversion back to the optically active substances. In most instances, the desired optical isomer is synthesîzed by means of stereospecific reactions, beginning with the appropriate stereoisomer of the desired starting material or through enantioselective reactions.
The compounds of the disclosure can also exist as tautomeric isomers in certain cases. Although only one tautomer may be depicted, ail such forms are contemplated within the scope of the disclosure. For example, ene-amine tautomers can exist for purine, pyrimidine, imidazole, guanidine, amidine, and tetrazole Systems and ail their possible tautomeric forms are within the scope of the disclosure.
Salts and Hydrates
Examples of physiologically or pharmaceutically acceptable salts of the compounds of the disclosure include salts derived from an appropriate base, such as, for example, an alkali métal (for example, sodium), an alkaline earth métal (for example, magnésium), ammonium and NXZ (wherein X is C1-C4 alkyl). Physiologically acceptable salts ofa hydrogen atom or an amino group include salts of organic carboxylic acids such as, for example, acetic, benzoic, lactic, fumaric, tartaric, maleic, malonic, malic, isethionic, lactobionic and succinic acids; organic sulfonic acids, such as, for example, methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids; and inorganic acids, such as, for example, hydrochloric, sulfuric, phosphoric and sulfamic acids. Physiologically acceptable salts of a compound of a hydroxy group include the anion of said compound in combination with a suitable cation such as, for example, Na+ and NX/ (wherein X is independently selected from H or a C1-C4 alkyl group).
For therapeutîc use, salts of active ingrédients ofthe compounds of the disclosure will typically be physiologically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base. However, salts of acids or bases which are not physiologically acceptable may also find use, for example, in the préparation or purification of a physiologically acceptable compound. AU salts, whether or not derived form a physiologically acceptable acid or base, are within the scope of the présent disclosure.
Métal salts typîcally are prepared by reacting the métal hydroxide with a compound of this disclosure. Examples of métal salts which are prepared in this way are salts containing Li+, Na+, and K+. A less soluble métal sait can be precipitated from the solution of a more soluble sait by addition of the suitable meta! compound.
In addition, salts may be formed from acid addition of certain organic and inorganic acids, e.g., HCl, HBr, H2SO4, H3PO4 or organic sulfonic acids, to basic centers, typîcally amines, or to acidic groups. Finally, it is to be understood that the compositions herein comprise compounds of the disclosure in their un-ionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates.
Also included within the scope of this disclosure are the salts of the parental compounds with one or more amino acids. Any of the naturel or unnatural amino acids are suitable, especially the naturally-occurring amino acids found as protein components, although the amino acid typîcally is one bearing a sîde chain with a basic or acidic group, e.g., lysine, arginine or glutamic acid, or a neutral group such as, for example, glycine, serine, threonine, alanine, isoleucine, or leucine.
Methods of Inhibition of HCV
Another aspect of the disclosure relates to methods of inhibiting the activity of HCV comprising the step of treating a sample suspected of containing HCV with a compound or composition of the disclosure.
The treating step of the disclosure comprises adding the compound of the disclosure to the sample or it comprises adding a precursor of the composition to the sample. The addition step comprises any method of administration as described above.
If desired, the activity of HCV after application of the compound can be observed by any method including direct and indirect methods ofdetecting HCV activity. Quantitative, qualitative, and semiquantitative methods of determining HCV activity are ail contemplated. Typîcally one of the screening methods described above are applied, however, any other method such as, for example, observation of the physiological properties of a living organism are also applicable.
Many organisais contain HCV. The compounds ofthis disclosure are usefiil in the treatment or prophylaxie of conditions associated with HCV activation in animais or in man.
However, in screening compounds capable of inhîbiring HCV activity it should be kept in mind that the results of enzyme assay s may not always correlate with cel! culture assays. Thus, a cell based assay should typically be the primary screening tool.
Pharmaceutical Formulations
The compounds of this disclosure are formulated with conventional carriers and excipients, which will be selected in accord with ordinary practice. Tablets will contain excipients, glidants, fil lers, binders and the like. Aqueous formulations are prepared in stérile form, and when intended for delivery by other than oral administration generally will be isotonie. Ail formulations will optionally contain excipients such as, for example, those set forth in the Handbook ofPharmaceutical Excipients (1986). Excipients include ascorbic acid and other antîoxidants, chelating agents such as, for example, EDTA, carbohydrates such as, for example, dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellulose, stearic acid and the like. The pH of the formulations ranges from about 3 to about 11, but is ordinarily about 7 to 10. Typically, the compound will be administered in a dose from 0.01 milligrams to 2 grams. In one embodiment, the dose will be from about 10 milligrams to 450 milligrams. It is contemplated that the compound may be administered once, twice or three rimes a day.
While it is possible for the active ingrédients to be administered alone, it may be préférable to présent them as pharmaceutical formulations. The formulations, both for veterinary and for human use, of the disclosure comprise at least one compound of the disclosure (herein referred to as active ingrédient), together with one or more acceptable carriers therefore and optionally other therapeutic ingrédients. The carrier(s) must be “acceptable” in the sense of being compatible with the other ingrédients of the formulation and physiologically innocuous to the récipient thereof.
The formulations include those suitable for the foregoing administration routes. The formulations may conveniently be presented in unit dosage form and may be prepared by any ofthe methods well known in the art ofpharmacy. Techniques and formulations generally are found in Remington’s Pharmaceutical Sciences (Mack Publishing Co., Easton, PA). Such methods include the step of bringing into association the active ingrédient with the carrier which constitues one or more accessory ingrédients. In general the formulations are prepared by uniformly and intîmately bringing into association the active ingrédient with liquid carriers or fmely divided solid carriers or both, and then, if necessary, shaping the product.
Formulations of the présent disclosure suitable for oral administration may be presented as discrète units such as, for example, capsules, cachets or tablets each containing a predetermined amount of the active ingrédient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid émulsion or a water-in-oil liquid émulsion. The active ingrédient may also be administered as a bolus, electuary or paste.
A tablet is made by compression or molding, optionally with one or more accessory ingrédients. Compressed tablets may be prepared by compressing in a suitable machine the active ingrédient în a free-flowing form such as, for example, a powder or granules, optionally mixed with a binder, fabricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered active ingrédient moistened with an inert liquid diluent. The tablets may optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingrédient therefrom.
For administration to the eye or other extemal tissues e.g., mouth and skin, the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w (including active ingredient(s) in a range between 0.1% and 20% in incréments of 0.1% w/w such as, for example, 0.6% w/w, 0.7% w/w, etc.), preferably 0.2 to 15% w/w and most preferably 0.5 to 10% w/w. When formulated in an ointment, the active ingrédients may be employed with either a parafïînic or a water-miscible ointment base. Altematively, the active ingrédients may be formulated in a cream with an oil-in-water cream base.
If desîred, the aqueous phase of the cream base may include, for example, at least 30% w/w of a polyhydric alcohol, i.e. an alcohol having two or more hydroxyl groups such as, for example, propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof. The topical formulations may desirably include a compound which enhances absorption or pénétration of the active ingrédient through the skin or other affected areas. Examples of such dermal pénétration enhancers include dïmethyl sulphoxide and related analogs.
The oily phase of the émulsions of this disclosure may be constituted from known ingrédients in a known manner. While the phase may comprise merely an emulsifier (otherwise known as an emulgent), it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilie emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the socalled emulsifying ointment base which forms the oily dispersed phase of the cream formulations.
Emulgents and émulsion stabilizers suitable for use in the formulation of the disclosure include Tween® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate.
The choîce of suitable oîls or fats for the formulation is based on achieving the desired cosmetic properties. The cream should preferably be a non-greasy, non-staining and washable product with suitable consistency to avoid leakage from tubes or other containers. Straight or branched chain, mono- or dibasic alkyl esters such as, for example, di-isoadipate, isocetyl stéarate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stéarate, 2-ethylhexyl palmitate or a blend of branched chain esters known as Crodamol CAP may be used, the last three being preferred esters. These may be used alone or in combination depending on the properties required. Altematively, high melting point lipids such as, for example, white soft paraflm and/or liquid paraffm or other minerai oils are used.
Pharmaceutical formulations according to the présent disclosure comprise one or more compounds of the disclosure together with one or more pharmaceutically acceptable carriers or excipients and optionally other therapeutic agents. Pharmaceutical formulations containing the active ingrédient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, émulsions, hard or soft capsules, syrups or élixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contaîn one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable préparation. Tablets containing the active ingrédient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as, for example, calcium or sodium carbonate, lactose, lactose monohydrate, croscarmellose sodium, povidone, calcium or sodium phosphate;
granulating and disintegrating agents, such as, for example, maize starch, or alginic acid; binding agents, such as, for example, cellulose, microcrystalline cellulose, starch, gelatin or acacia; and lubricating agents, such as, for example, magnésium stéarate, stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as, for example, glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
Formulations for oral use may be also presented as hard gelatin capsules where the active ingrédient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingrédient is mixed with water or an oil medium, such as, for example, peanut oil, liquid paraffin or olive oil.
Aqueous suspensions of the disclosure contain the active materials in admixture with excipients suitable for the manufacture ofaqueous suspensions. Such excipients include a suspending agent, such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as, for example, a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stéarate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives such as, for example, ethyl or n-propyl p-hydroxy-benzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as, for example, sucrose or saccharin.
Oil suspensions may be formulated by suspending the active ingrédient in a vegetable oil, such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a minerai oil such as, for example, liquid paraffin. The oral suspensions may contain a thickening agent, such as, for example, beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as, for example, those set forth above, and flavoring agents may be added to provide a palatable oral préparation. These compositions may be preserved by the addition of an antioxidant such as, for example, ascorbic acid.
Dispersible powders and granules of the disclosure suitable for préparation of an aqueous suspension by the addition of water provide the active ingrédient in admixture with a dispersing or wetting agent, a suspending agent, and one or more préservâtives. Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be présent.
The pharmaceutical compositions of the disclosure may also be in the form of oil-inwater émulsions. The oily phase may be a vegetable oïl, such as, for exampte, olive oil or arachis oil, a minerai oil, such as, for example, liquid parafïîn, or a mixture of these. Suitable emulsifying agents include naturel ly-occurring gums, such as, for example, gum acacia and gum tragacanth, naturally occurring phosphatîdes, such as, for example, soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as, for example, sorbitan monooleate, and condensation products of these partial esters with ethylene oxide, such as, for example, polyoxyethylene sorbitan monooleate. The émulsion may also contain sweetening and flavoring agents. Syrups and élixirs may be formulated with sweetening agents, such as, for example, glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
The pharmaceutical compositions of the disclosure may be in the form of a stérile injectable préparation, such as, for example, a stérile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which hâve been mentioned above. The stérile injectable préparation may also be a stérile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as, for example, a solution in 1,3-butane-diol or prepared as a lyophilized powder. Among the acceptable vehîcles and solvents that may be employed are water, Ringer’s solution and isotonie sodium chloride solution. In addition, stérile fixed oîls may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as, for example, oleic acid may likewise be used in the préparation of injectables.
The amount of active ingrédient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral administration to humans may contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions (weightweight). The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion may contain from about 3 to 500 pg of the active ingrédient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.
Formulations suitable for administration to the eye include eye drops wherein the active ingrédient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingrédient. The active ingrédient is preferably présent in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% particularly about l .5% w/w.
Formulations suitable for topical administration in the mouth include lozenges comprising the active ingrédient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingrédient in an inert basis such as, for example, gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingrédient in a suitable liquîd carrier.
Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
Formulations suitable for intrapulmonary or nasal administration hâve a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in incréments microns such as, for example, 0.5, 1,30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations include aqueous or oily solutions of the active ingrédient. Formulations suitable for aérosol or dry powder administration may be prepared according to conventional methods and may be delivered with other therapeutic agents such as, for example, compounds heretofore used in the treatment or prophylaxis of conditions associated with HCV activity.
Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations contaîning in addition to the active ingrédient such carriers as are known in the art to be appropriate.
Formulations suitable for parentéral administration include aqueous and non-aqueous stérile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutés which render the formulation isotonie with the blood of the intended récipient; and aqueous and non-aqueous stérile suspensions which may include suspending agents and thickening agents.
The formulations are presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a ffeeze-dried (lyophilized) condition requiring only the addition of the stérile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from stérile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingrédient.
It should be understood that in addition to the ingrédients particularly mentioned above the formulations ofthis disclosure may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
The disclosure further provides veterinary compositions comprising at least one active ingrédient as above defîned together with a veterinary carrier therefore.
Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingrédient. These veterinary compositions may be administered orally, parenterally or by any other desired route.
Compounds of the disclosure can also be formulated to provide controlled release of the active ingrédient to allow less frequent dosing or to improve the pharmacokinetic or toxicity profile of the active ingrédient. Accordingly, the disclosure also provides compositions comprising one or more compounds of the disclosure formulated for sustained or controlled release.
Effective dose of active ingrédient dépends at least on the nature of the condition being treated, toxicity, whether the compound is being used prophylactically (lower doses), the method of delivery, and the pharmaceutical formulation, and will be determined by the clinician using conventional dose escalation studies.
In one embodiment, the active ingrédient (i.e., one or more compounds as described herein) or pharmaceutical composition comprising the active ingrédient are effective in treating one or more of génotype 1 HCV infected subjects, génotype 2 HCV infected subjects, génotype 3 HCV infected subjects, génotype 4 HCV infected subjects, génotype 5 HCV infected subjects, and/or génotype 6 HCV infected subjects. In one embodiment, the active ingrédient or pharmaceutical composition comprising the active ingrédient are effective in treating génotype 1 HCV infected subjects, including génotype la and/or génotype lb. In another embodiment, the active ingrédient or pharmaceutical composition comprising the active ingrédient are effective in treating génotype 2 HCV infected subjects, including génotype 2a, génotype 2b, génotype 2c and/or génotype 2d. In another embodiment, the active ingrédient or pharmaceutical composition comprising the active ingrédient are effective in treating génotype 3 HCV infected subjects, including génotype 3a, génotype 3 b, génotype 3 c, génotype 3d, génotype 3e and/or génotype 3 f. In another embodiment, the active ingrédient or pharmaceutical composition comprising the active ingrédient are effective in treating génotype 4 HCV infected subjects, including génotype 4a, génotype 4b, génotype 4c, génotype 4d, génotype 4e, génotype 4f, génotype 4g, génotype 4h, génotype 4i and/or génotype 4j. In another embodiment, the active ingrédient or pharmaceutical composition comprising the active ingrédient are effective in treating génotype 5 HCV infected subjects, including génotype 5a. In another embodiment, the active ingrédient or pharmaceutical composition comprising the active ingrédient effective in treating génotype 6 HCV infected subjects, including génotype 6a.
In some embodiments, the active ingrédient or pharmaceutical composition comprising the active ingrédient is administered, either alone or in combination with one or more therapeutic agent(s) for treating HCV (such as a HCV NS3 protease inhibitor or an inhibitor of HCV NS5B polymerase), for about 24 weeks, for about 16 weeks, or for about 12 weeks, or less. In further embodiments, the active ingrédient or pharmaceutical composition comprising the active ingrédient is administered, either alone or in combination with one or more therapeutic agent(s) for treating HCV (such as a HCV NS3 protease inhibitor or an inhibitor of HCV NS5B polymerase), for about 24 weeks or less, about 22 weeks or less, about 20 weeks or less, about 18 weeks or less, about 16 weeks or less, about 12 weeks or less, about 10 weeks or less, about 8 weeks or less, or about 6 weeks or less or about 4 weeks or less. The active ingrédient or pharmaceutical composition comprising the active ingrédient may be administered once daily, twice daily, once every other day, two times a week, three times a week, four times a week, or five times a week.
In further embodiments, a sustained virologie response is achieved at about 4 weeks, 6 weeks, 8 weeks, 12 weeks, or 16 weeks, or at about 20 weeks, or at about 24 weeks, or at about 4 months, or at about 5 months, or at about 6 months, or at about 1 year, or at about 2 years.
Routes of Administration
One or more compounds of the disclosure (herein referred to as the active ingrédients) are administered by any route appropriate to the condition to be treated. Suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parentéral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and épidural), and the like. It will be appreciated that the preferred route may vary with for example the condition ofthe récipient. An advantage ofthe compounds ofthis disclosure is that they are oralty bioavailable and can be dosed orally.
HCV Combination Therapy
In another embodiment, non-limiting examples of suitable combinations include combinations of one or more compounds of formula (I) and (Al-A4) with one or more interferons, ribavirin or its analogs, HCV NS3 protease inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, nucleoside or nucléotide inhibitors of HCV NS5B polymerase, non-nucleoside inhibitors of HCVNS5B polymerase, HCV NS5A inhibitors, TLR-7 agonists, cyclophillin inhibitors, HCV IRES inhibitors, pharmacokinetic enhancers, and other drugs or therapeutic agents for treating HCV.
More specifically, one or more compounds of the présent as described herein may be combined with one or more compounds selected from the group consisting of
1) interferons, e.g., pegylated rIFN-alpha 2b (PEG-Intron®), pegylated rIFN-alpha 2a (Pegasys®), rIFN-alpha 2b (Intron® A), rIFN-alpha 2a (Roferon®-A), interferon alpha (MOR-22, OPC-18, Alfaferone®, Alfanative®, Multiferon®, subalîn), interferon alfacon-1 (Infergen®), interferon a!pha-nl (Wellferon), interferon alpha-n3 (Alferon®), interferon-beta (Avonex®, DL-8234), interferon-omega (oméga DUROS®, Bîomed® 510), albinterferon alpha-2b (Albuferon®) IFN alpha-2b XL, BLX-883 (Locteron®), DA-3021, glycosylated interferon alpha-2b (AVI-005), PEG-Infergen, PEGylated interferon lambda-1 (PEGylated IL-29), and belerofon®;
2) ribavirin and its analogs, e.g., ribavirin (Rebetol®, Copegus®), and taribavirin (Viramidine®);
3) HCV NS3 protease inhibitors, e.g., boceprevir (SCH-503034, SCH-7), telaprevir (VX-950), TMC435350, BI-1335, BI-1230, MK-7009, VBY-376, VX-500, GS-9256, GS9451, BMS-605339, PHX-1766, AS-101, YH-5258, YH553O, YH5531, ABT-450, ACH1625, ΓΓΜΝ-191, AT26893, MK5172, MK6325, and MK2748;
4) alpha-glucosidase 1 inhibitors, e.g., celgosivir (MX-3253), Miglitol, and UT-23IB;
5) hepatoprotectants, e.g., emericasan (IDN-6556), ME-3738, GS-9450 (LB-84451), silibilin, and MitoQ;
6) nucleoside or nucléotide inhibitors of HCV NS5B polymerase, e.g., RI 626, R7128 (R4048), IDX184, IDX-102, BCX-4678, valopicitabine (NM-283), MK-0608, sofosbuvir (GS-7977 (formerly PSI-7977)), VLX-135 (formerly ALS-2200), and INX-189 (now BMS986094);
7) non-nucleoside inhibitors of HCV NS5B polymerase, e.g., PF-868554, VCH-759, VCH-916, JTK-652, MK-3281, GS-9190, VBY-708, VCH-222, A848837, ANA-598, GL60667, GL59728, A-63890, A-48773, A-48547, BC-2329, VCH-796 (nesbuvir), GSK625433, BILN-1941, XTL-2125, ABT-072, ABT-333, GS-9669, PSI-7792, and GS9190;
8) HCV NS5A inhibitors, e.g., AZD-2836 (A-831), BMS-790052, ACH-3102, ACH2928, MK8325, MK4882, MK8742, PSI-461, IDX719, GS-5885, and A-689;
9) TLR-7 agonists, e.g., imiquimod, 852A, GS-9524, ANA-773, ANA-975 (isatoribine), AZD-8848 (DSP-3025), and SM-360320;
10) cyclophillin inhibitors, e.g., DEBIO-025, SCY-635, and NIM811;
11) HCV IRES inhibitors, e.g., MCI-067;
12) pharmacokinetic enhancers, e.g., BAS-100, SPI-452, PF-4194477, TMC-41629, GS-9350 (cobicistat), GS-9585, and roxythromycin; and
13) other drugs for treating HCV, e.g., thymosin alpha 1 (Zadaxin), nitazoxanide (Alinea, NTZ), BIVN-401 (virostat), PYN-17(altirex), KPE02003002, actilon(CPG-lOlOl), GS-9525, KRN-7000, civacir, GI-5005, XTL-6865, BIT225, PTX-111, ΓΓΧ2865, TT-033Î, ANA 971, NOV-205, tarvacîn, EHC-18, VGX-410C, EMZ-702, AVI4065, BMS-650032, BMS-791325, Bavituximab, MDX-1106 (ONO-4538), Oglufanide, and VX-497 (merimepodib).
In yet another embodiment, the présent application discloses pharmaceutical compositions comprising a compound as described herein, or a pharmaceutically acceptable sait, solvaté, and/or ester thereof in combination with at least one additional therapeutic agent, and a pharmaceutically acceptable carrier or excipient.
More specifically, the additional therapeutic agent may be combined with one or more compounds selected from the group consisting of non-nucleoside inhibitors of HCV NS5B polymerase (ABT-072 and ABT-333), HCV NS5A inhibitors (ACH-3102 and ACH-2928) and HCV NS3 protease inhibitors (ABT-450 and ACH-125).
In another embodiment, the therapeutic agent used in combination with the pharmaceutical compositions as described herein can be any agent having a therapeutic effect when used in combination with the pharmaceutical compositions as described herein. For example, the therapeutic agent used in combination with the pharmaceutical compositions as described herein can be interferons, ribavirin analogs, NS3 protease inhibitors, NS5B polymerase inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, non-nucleoside inhibitors of HCV, and other drugs for treating HCV.
In another embodiment, the additional therapeutic agent used in combination with the pharmaceutical compositions as described herein is a cyclophillin inhibitor, including for example, a cyclophilin inhibitor disclosed in WO/2013/185093. Non-limiting examples include one or more compounds selected from the group consisiting of;
thereof.
In another embodiment, the additional therapeutic agent used in combination with the pharmaceutical compositions as described herein is a non-nucleoside inhibitor of HCV NS5B polymerase. A non-limiting example includes GS-9669.
Examples of additional anti-HCV agents which can be combined with the compositions provided herein include, without limitation, the following:
A. interferons, for example, pegylated rIFN-alpha 2b (PEG-Intron), pegylated rIFN-alpha 2a (Pegasys), rIFN-alpha 2b (lntron A), rIFN-alpha 2a (Roferon-A), interferon alpha (MOR-22, OPC-18, Alfaferone, Alfanative, Multiferon, subalîn), interferon alfacon-1 (Infergen), interferon alpha-nl (Wellferon), interferon alpha-n3 (Alferon), interferon-beta (Avonex, DL-8234), interferon-omega (oméga DUROS, Biomed 510), albinterferon alpha-2b (Albuferon), IFN alpha XL, BLX-883 (Locteron), DA-3021, glycosylated interferon a!pha-2b (AVI-005), PEG-Infergen, PEGylated interferon lambda (PEGylated IL-29), or belerofon, IFN alpha-2b XL, rIFN-alpha 2a, consensus IFN alpha, infergen, rebif, pegylated IFN-beta, oral interferon alpha, feron, reaferon, intermax alpha, r-IFN-beta, and infergen + actimmuneribavirin and ribavirin analogs, e.g., rebetol, copegus, VX-497, and viramidine (taribavirin);
B. NS5A inhibitors, for example, Compound B (described below), Compound C (described below), ABT-267, Compound D (described below), JNJ-47910382, daclatasvir (BMS-790052), ABT-267, MK-8742, EDP-239, IDX-719, PPI-668, GSK-2336805, ACH3102, A-831, A-689, AZD-2836 (A-831), AZD-7295 (A-689), and BMS-790052;
C. NS5B polymerase inhibitors, for example, Compound E (described below), Compound F (described below), ABT-333, Compound G (described below), ABT-072, Compound H (described below), tegobuvir (GS-9190), GS-9669, TMC647055, setrobuvir (ANA-598), filibuvir (PF-868554), VX-222, IDX-375, IDX-184, IDX-102, BI-207127, valopicitabine (NM-283), PSI-6130 (R1656), PSI-7851, BCX-4678, nesbuvir (HCV-796), BILB 1941, MK-0608, NM-107, R7128, VCH-759, GSK625433, XTL-2125, VCH-916,
JTK-652, MK-3281, VBY-708, A848837, GL59728, A-63890, A-48773, A-48547, BC-2329, BMS-791325, and BILB-1941;
D. NS3 protease inhibitors, for example, Compound I, Compound J, Compound K, ABT-450, Compound L (described below), sîmeprevir (TMC-435), boceprevir (SCH503034), narlaprevir (SCH-900518), vaniprevir (MK-7009), MK-5172, danoprevir (ITMN191), sovaprevir (ACH-1625), neceprevir (ACH-2684), Telaprevir (VX-950), VX-813, VX500, faldaprevir (BI-201335), asunaprevir (BMS-650032), BMS-605339, VBY-376, PHX1766, YH5531, BILN-2065, and BILN-2061;
E. alpha-glucosidase 1 inhibitors, for example, celgosivir (MX-3253), Miglitol, and UT-231B;
F. hepatoprotectants, e.g., IDN-6556, ME 3738, MitoQ, and LB-84451 ;
G. non-nucleoside inhibitors of HCV, e.g., benzimidazole dérivatives, benzo1,2,4-thiadîazine dérivatives, and phenylalanîne dérivatives; and
H. other anti-HCV agents, e.g., zadaxin, nitazoxanide (alinea), BIVN-401 (virostat), DEBIO-025, VGX-410C, EMZ-702, AVI4065, bavituximab, oglufanide, PYN-17, KPE02003002, actilon (CPG-10101), KRN-7000, civacir, GI-5005, ANA-975, XTL-6865, ANA 971, NOV-205, tarvacin, EHC-18, and NIM811.
Compound B is an NS5A inhibitor and is represented by the following chemical structure:
Compound C is an NS5A inhibitor and is represented by the following chemical
Compound D is an NS5 A inhibitor and is represented by the following chemical structure:
See U.S. Publication No. 2013/0102525 and référencés thereîn.
Compound E is an NS5B Thumb II polymerase inhibitor and is represented by the following chemical structure:
Compound F is a nucléotide inhibitor prodrug designed to inhibit réplication of viral RNA by the HCV NS5B polymerase, and is represented by the following chemical structure:
Compound G is an HCV polymerase inhibitor and is represented by the following structure:
See U.S. Publication No. 2013/0102525 and référencés therein.
Compound H is an HCV polymerase inhibitor and is represented by the following structure:
NHSOjCHa
See U.S. Publication No. 2013/0102525 and référencés thereîn.
Compound I is an HCV protease inhibitor and is represented by the following chemical structure:
See PCT/US2013/049119, filed July 2,2013, and référencés therein.
Compound J is an HCV protease inhibitor and is represented by the following chemical structure:
Compound K is an HCV protease inhibitor and is represented by the following chemical structure:
Compound L is an HCV protease inhibitor and is represented by the following chemical structure:
See U.S. Publication No. 2013/0102525 and references therein.
In one embodîment, the additional therapeutic agent used in combination with the pharmaceutical compositions as described herein is a HCV NS3 protease inhibitor. Nonlimiting examples include one or more compounds selected from the group consisiting of:
In another embodiment, the présent application provides for a method of treating hepatitis C in a human patient in need thereof comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition as described herein and an additional therapeutic selected from the group consisting of pegylated rIFN-alpha 2b, pegylated rIFN-alpha 2a, rIFN-alpha 2b, IFN a!pha-2b XL, rIFN-alpha 2a, consensus IFN alpha, infergen, rebif, locteron, AVI-005, PEG-infergen, pegylated IFN-beta, oral interferon alpha, feron, reaferon, intermax alpha, r-IFN-beta, infergen + actimmune, IFN-omega with DUROS, albuferon, rebetol, copegus, levovirin, VX-497, viramidine (taribavirin), A-831, A689, NM-283, valopicitabine, R1626, PSI-6130 (R1656), HCV-796, BILB 1941, MK-0608, NM-107, R7128, VCH-759, PF-868554, GSK625433, XTL-2125, SCH-503034 (SCH-7), VX-950 (Telaprevir), ΓΓΜΝ-191, and BILN-2065, MX-3253 (celgosivir), UT-231B, IDN6556, ME 3738, MitoQ, and LB-84451, benzimidazole dérivatives, benzo-l,2,4-thiadiazine dérivatives, and phenylalanîne dérivatives, zadaxin, nitazoxanide (alinea), BIVN-401 (virostat), DEBIO-025, VGX-410C, EMZ-702, AVI4065, bavituximab, oglufanide, PYN-17, KPE02003002, actilon (CPG-10101), KRN-7000, civacir, GI-5005, ANA-975 (isatoribine), XTL-6865, ANA 971, NOV-205, tarvacin, EHC-18, and NIM811 and a pharmaceutically acceptable carrier or excipient.
In another embodiment is provided a pharmaceutical composition comprising a compound of formula (I) as described herein and sofosbuvir and/or GS-5885 and optionally an interferon or ribavirin.
It is contemplated that additional therapeutic agents will be administered in a manner that is known in the art and the dosage may be selected by someone of skill in the art. For example, additional therapeutic agents may be administered in a dose from about 0.01 milligrams to about 2 grams per day.
Métabolites of the Compounds
Also falling within the scope of this disclosure are the in vivo metabolic products of the compounds described herein. Such products may resuit for example from the oxidation, réduction, hydrolysis, amidation, estérification and the like ofthe administered compound, primarily due to enzymatic processes. Accordingly, the disclosure includes compounds produced by a process comprising contactîng a compound of this disclosure with a mammal for a period of time sufïïcient to yield a metabolic product thereof. Such products typically are identified by preparing a radiolabelled (e.g., C14 or H^) compound of the disclosure, administering it parenterally in a détectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as, for example, rat, mouse, guinea pig, monkey, or to man, allowing sufïïcient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the métabolite). The métabolite structures are determined in conventional fashion, e.g., by MS or NMR analysis. In general, analysis of métabolites is done in the same way as conventional drug metabolism studies well-known to those skilled in the art. The conversion products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the disclosure even if they possess no HCV -inhibitory activity of their own.
Methods for determining stability of compounds in surrogate gastrointestinal sécrétions are known.
Exemplary Methods of Making the Compounds
The disclosure also relates to methods of making the compositions of the disclosure. The compositions are prepared by any of the applicable techniques of organic synthesis. Many such techniques are well known in the art. However, many of the known techniques are elaborated in Compendium of Organic Synthetic Methods (John Wiley & Sons, New York), Vol. l.IanT. Harrison and Shuyen Harrison, 1971; Vol. 2, IanT. Harrisonand Shuyen Harrison, 1974; Vol 3, Louis S. Hegedus and Leroy Wade, 1977, Vol. 4, Leroy G. Wade, Jr., 1980; Vol. 5, Leroy G. Wade, Jr., 1984; and Vol. 6, Michael B. Smith; as well as March, J., Advanced Organic Chemistry, Third Edition, (John Wiley & Sons, New York, 1985), Comprehensive Organic Synthesis. Selectivity, Strategy & Efïïciency in Modem Organic Chemistry. In 9 Volumes, Barry M. Trost, Editor-in-Chief (Pergamon Press, New
York, 1993 printing). Other methods suitable for preparing compounds ofthe disclosure are described in International Patent Application Publication Number WO 2006/020276.
A number of exemplary methods for the préparation of the compositions of the disclosure are provided in the schemes and examples below. These methods are intended to îllustrate the nature of such préparations and are not intended to limit the scope of applicable methods.
Generally, the reaction conditions such as, for example, température, reaction time, solvents, work-up procedures, and the like, will be those common in the art for the particular réaction to be performed. The cited reference material, together with material cited therein, contains detailed descriptions of such conditions. Typically the températures will be -100°C to 200°C, solvents will be aprotic or protic, and reaction times will be 10 seconds to 10 days. Work-up typically consists of quenchîng any unreacted reagents followed by partition between a water/organic layer System (extraction) and separating the layer containing the product.
Oxidation and réduction reactions are typically carried out at températures near room température (about 20°C), although for meta! hydride réductions frequently the température is reduced to 0°C to -100°C, solvents are typically aprotic for réductions and may be either protic or aprotic for oxidations. Reaction times are adjusted to achieve desired conversions.
Condensation reactions are typically carried out at températures near room température, although for non-equilibrating, kinetically controlled condensations reduced températures (0°C to -100°C) are also common. Solvents can be either protic (common in equilibrating reactions) or aprotic (common in kinetically controlled reactions).
Standard synthetic techniques such as, for example, azeotropic removal of reaction by-products and use of anhydrous reaction conditions (e.g., inert gas environments) are common in the art and will be applied when applicable.
The terrns “treated”, “treating”, “treatment”, and the like, when used in connection with a chemical synthetic operation, mean contacting, mixing, reacting, allowing to react, bringing into contact, and other terrns common in the art for indicating that one or more chemical entities is treated in such a manner as to convert it to one or more other chemical entities. This means that “treating compound one with compound two” is synonymous with “allowing compound one to react with compound two”, “contacting compound one with compound two”, “reacting compound one with compound two”, and other expressions common in the art of organic synthesis for reasonably indicating that compound one was “treated”, “reacted”, “allowed to react”, etc., with compound two. For example, treating indicates the reasonable and usual manner in which organic chemicals are allowed to react. Normal concentrations (0.0IM to 10M, typically 0.1M to IM), températures (-100°C to 250°C, typically -78°C to I50°C, more typically -78°C to 100°C, still more typically 0°C to 100°C), reaction vessels (typically glass, plastic, métal), solvents, pressures, atmosphères (typically air for oxygen and water insensitive reactions or nitrogen or argon for oxygen or water sensitive), etc., are intended unless otherwise indicated. The knowledge of similar reactions known in the art of organic synthesis is used in selecting the conditions and apparatus for “treating” in a given process. In particular, one of ordinary skill in the art of organic synthesis selects conditions and apparatus reasonably expected to successfully carry out the chemical reactions of the described processes based on the knowledge in the art.
Modifications of each of the exemplary schemes and in the Examples (hereafter “exemplary schemes”) leads to various analogs of the spécifie exemplary materials produce. The above-cited citations describing suitable methods of organic synthesis are applicable to such modifications.
In each of the exemplary schemes it may be advantageous to separate reaction products from one another and/or from starting materials. The desired products of each step or sériés of steps is separated and/or purified (hereinafter separated) to the desired degrec of homogeneity by the techniques common in the art. Typically such séparations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium, and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and préparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
Another class of séparation methods involves treatment of a mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the Jike. Such reagents include adsorbents or absorbents such as, for example, activated carbon, molecular sieves, ion exchange media, or the like. Altematively, the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as, for example, antibodies, binding proteins, sélective chelators such as, for example, crown ethers, liquid/liquid ion extraction reagents (LIX), or the like.
Sélection of appropriate methods of séparation dépends on the nature of the materials involved. For example, boiling point, and molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic and basic media in multiphase extraction, and the like. One skilled in the art wil! apply techniques most likely to achieve the desired séparation.
A single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as, for example, formation of diastereomers using optically active resolving agents (Stereochemistry of Carbon Compounds, (1962) by E. L. Eliel, McGraw Hill; Lochmuller, C. H., (1975) J. Chromatogr., 113, 3) 283-302). Racemic mixtures of chiral compounds of the disclosure can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and séparation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, séparation of the diastereomers, and conversion to the pure stereoîsomers, and (3) séparation of the substantially pure or enriched stereoîsomers directly under chiral conditions.
Under method (1), diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as, for example, brucine, quinine, ephedrine, strychnine, ot-methyl-βphenylethylamine (amphétamine), and the like with asymmetric compounds bearing acidic functionality, such as, for example, carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography. For séparation of the optica! isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as, for example, camphorsulfonic acid, tartaric acid, mandelîc acid, or lactic acid can resuit in formation of the diastereomeric salts.
Altematively, by method (2), the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and Wilen, S. (1994) Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., p. 322). Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as, for example, menthyl dérivatives, followed by séparation of the diastereomers and hydrolysis to yield the free, enantiomerically enriched substrate. A method of determining optical purity invokes making chiral esters, such as, for example, a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, a-methoxy-a-(trifluoromethyl)phenyl acetate (Jacob ΠΙ. (1982) J. Org. Chem. 47:4165), of the racemic mixture, and analyzîng the NMR spectrum for the presence ofthe two atropisomeric diastereomers. Stable diastereomers ofatropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for séparation of atropisomeric naphthyl-isoquinolines (Hoye, T., WO 96/15111). By method (3), a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase (Chiral Liquid Chromatography (1989) W. J. Lough, Ed. Chapman and Hall, New York; Okamoto, (1990) J. of Chromatogr. 513:375-378). Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molécules with asymmetric carbon atoms, such as, for example, optical rotation and circular dichroism.
Schemes and Exemples
General aspects of these exemplary methods are described below and in the Examples. Each of the products of the following processes is optionally separated, isolated, and/or purified prior to its use in subséquent processes.
A number of exemplary methods for the préparation of compounds of the disclosure are provided herein, for example, in the Examples below. These methods are intended to illustrate the nature of such préparations and are not intended to limit the scope of applicable methods. Certain compounds ofthe disclosure can be used as intermediates for the préparation of other compounds of the disclosure. In the exemplary methods described herein, the fragment E-V- can also be written as R9-. PG représente a protecting group common for the given fùnctional group that it is attached. The installation and removal of the protecting group can be accomplished using standard techniques, such as those described in Wuts, P. G. M., Greene, T. Protecnve Groups in Organic Synthesis, 4th ed.; John Wiley & Sons, Inc.: Hoboken, New Jersey, 2007.
Scheme 1. Représentative synthesis of E-V-C(=O)-P-W-P-C(=O)-V-E o
''cAci
H2N-V-C(=O)-P-W-P-C(=O)-VÆ ---------1a 2 Ά
H2N-VX?(=0)-P-W-P-C(=O)-V-NH2 ---------1c c>
^-NH-V-C(=O)-P-W-P-C(=O)-V-E °x ’» oo
VNH-V-C(=O)-P-W-P-C(=O)-V-NH-# oo \ 1d/
Scheme 1 shows a general synthesis of an E-V-C(=O)-P-W-P-C(=O)-V-E molécule of the disclosure wherein, for illustrative purposes, E is methoxycarbonylamîno. The treatment of either la or le with one or two équivalents respectively of methyl chloroformate under basic conditions (e.g. sodium hydroxide) provides the molécule 1b or Id.
Scheme 2. Représentative synthesis of E-V-C(=O)-P-W-P-C(=O)-V-E
Ε-ν-0(=Ο}-Ρ-νν-\Ί hr 2c oA
V-E E
2e V-E
2e oA
V-E
Scheme 2 shows a general synthesis of an E-V-C(=O)-P-W-P-C(=O)-V-E molécule 10 of the disclosure wherein, for illustrative purposes, P is pyrrolidine. Coupîing of amine 2a with acid 2b is accomplished using a peptide coupîing reagent (e.g. HATU) to afford 2c. Altematively, amine 2d is coupled with two équivalents of 2b under sîmilar conditions to provide 2e. Altematively, amine 2d is reacted with two équivalents of 2b* directly to provide 2e where E* is a leaving group such as hydroxybenztriazole, para-nitrophenol or the like making the structure 2b’ an activated ester.
Scheme 3. Représentative synthesis of R’-V-C(=O)-P-R2
E-V-C(=O)-P-W—\Ί
N^ 3a H
HO ^-V-NH-PG O
3b
E-V-C(=O)-P-W—Z]
Iv 3c oX V-NH-PG
PG-HN-V-C(=O)-P-W—(Ί
3d H
HO y™ o 3e
PG-HN-V-C(=O|-P-W-(] N^
PG-HN-V-C(=O)-P-W -<]
3d H
HO + V-NH-PG O
3b
PG-HN-P-W—
3h H
HO y^ o 3e
PG-HN-V-C(=O)-P-W— 39 oX
V-NH-PG
PG-HN-P-W -(] 3i oX
V-E
PG-HN-P-W—( ]
3h H
HO ^-V-NH-PG O
3b
PG-HN-P-W -(Ί * oX V-NH-PG
PG-HN-W—ς]
3k H
HO y^ o 3e
PG-HN-W—ζ] 31 oX
V-E
HO V-NH-PG O
3b
PG-HN-W—<]
3k H
PG-HN-W—
N^ 3m oX V-NH-PG
Scheme 3 shows a general synthesis of an R’-V-C(=O)-P-R2 intermediate wherein, for illustrative purposes, P is pyrrolidine, R1 is a generic group that is depicted as either -E or 5 a amino protecting group, and R2 is a generic group that is depicted as -W-P-C(=O)-V-E, W-P-C(=O)-V-NH-PG, -W-P-NH-PG, or -W-NH-PG. Coupling of amine 3a (or 3d, 3h,
3k) with acid 3b or 3e is accomplished using a peptide coupling reagent (e.g. HATU) to afford 3c (or 3f, 3g, 3i, 3j, 31,3m) respectively.
Scheme 4. Représentative synthesis of E-V-C^OJ-R*
H2N-V-C(=0)-P-W-P-C(=O)-V-NH-PG - 0 0 ^-NH-V-C(=O}-P-W-P-C(=O)-V-NH-PG
4a o °\ 4b
O
H2N-V-C(=O)-P-W-P-PG ^-NH-V-C(=O)-P-W-P-PG
4c o °\ «
0
H2N-V-C(=O)-P-W-PG y-NH-V-C(=O)-P-W-PG
4e o ° 4!
O
^-NH-V-C(=O)-P-FG
4g 0 O 4b
o
H2N-V-C(=O)-O-PG VnH-V-C(=OEO-PG
41 O « \ 4
Scheme 4 shows a general synthesis of an E-V-CX^OJ-R1 intermediate wherein, for illustrative purposes, E is methoxycarbonylamino and R1 is a generic group that is depicted 5 as either -P-W-P-C(=O)-V-NH-PG, -P-W-P-PG, -P-W-PG, -P-PG, or -O-PG Treatment of 4a (or 4c, 4e, 4g, 4i) with methyl chloroformate under basic conditions (e.g. sodium hydroxide) provides the molécule 4b (or 4d, 4f, 4h, 4j).
Scheme 5. Représentative synthesis of R’-P-W-P-R2
Scheme 5 shows a general synthesis of an R’-P-W-P-R2 intermediate of the disclosure wherein, for illustrative purposes, R* and R2 are independent protecting groups and 5 W is a two aromatic ring unit constructed via a transition métal mediated cyclization.
Alkylation of phénol 5b with an alkyl bromide, such as 5a, provides the ether 5c. Cyclization of the aromatic rings in the presence of a palladium catalyst provides the compound 5d. Treatment of 5d with CuBq provides the α-haloketone 5e, which provides 5f upon addition of an acid under basic conditions (e.g. EfcN). Reaction of 5f with an amine or amine sait (e.g.
ammonium acetate) affords the imidazole containing molécule 5g. Oxidation of 5g, 5i, or 51 can be accomplished by heating in the presence of MnOj to provide 5h, 5j, or 5m, respectively. Conversion of 5g or 5h with a palladium catalyst, such as Pdzdbai and X-Phos, and a boron source such as bis(pinacolato)diboron provides the boronic ester Si or 5j. The boronic ester is coupled with an appropriate coupling partner (e.g. 5k) using a palladium catalyst, such as Pd(PPh3)-i or PdCh(dppf), to afford SI or 5m. For each transition métal mediated cross-coupling reaction, the rôles of the nucleophile and electrophile can be 5 reversed to provide the same coupling product. Other transition métal mediated cross couplings that enable the construction of W, but employ alternative coupling partners and reagents, include, but are not limited to, theNegishi, Kumada, Stille, and Ullman couplings. For the préparation of altemate two aromatic ring containing W groups, this general scheme can be applied through the appropriate choice of the starting reagents.
Scheme 6. Représentative synthesis of R’-P-W-P-R2
O
O
6f/g
Scheme 6 shows a general synthesis of an R’-P-W-P-R2 intermediate of the disclosure wherein, for illustrative purposes, R* and R2 are independent protecting groups and W is a two aromatic ring unit constructed via a transition métal mediated cyclization. Treatment of 5d with an activated vinyl reagent (e.g. potassium vinyltrifluoroborate) in the presence of a palladium catalyst (e.g. palladium acetate and S-Phos) provides the vinyl compound 6a. Conversion to the corresponding α-halo ketone can be accomplished by bromination with N-bromosuccinimide, followed by oxidation with MnO2. Displacement of the α-halo ketone proceeds by the addition of an acid under basîc conditions (e.g. Et3N). Bromination of 6d proceeds upon treatment with pyridinium tribromide, and is followed by the addition of a second acid under basic conditions to provide the diester 6e. Reaction of 6e with an amine or amine sait (e.g. ammonium acetate) afFords the imidazole containing molécule 6f. Oxidation of6f can be accomplished in the presence of MnO2 to provide 6g.
Scheme 7. Représentative synthesis of E-V-C(=O)-P-W-P-R
Scheme 7 shows a general synthesis of an E-V-C(=O)-P-W-P-R intermediate of the disclosure wherein, for illustrative purposes, R is a protecting group and W is a two aromatic ring unit. Displacement of the α-halo ketone 6b proceeds by the addition of an acid under basic conditions (e.g. EhN)· Bromination of 7b proceeds upon treatment with pyridinium 5 tribromide, and is followed by the addition of a second acid under basic conditions to provide the diester 7c. Reaction of 7c with an amine or amine sait (e.g. ammonium acetate) afîords the imidazole containing molécule 7d. Oxidation of 7d can be accomplished in the presence of MnO2 to provide 7e.
Scheme 8. Représentative synthesis of R-P-W-P-C(=O)~V-E
Scheme 8 shows a general synthesis of an E-V-C(=O)-P-W-P-R intermediate of the disclosure wherein, for illustrative purposes, Ris a protecting group and W is a two aromatic ring unît. Displacement of the α-halo ketone 6d proceeds by the addition of an acid under basic conditions (e.g. Et3N). Reaction of 8a with an amine or amine sait (e.g. ammonium 15 acetate) afîords the imidazole containing molécule 8b. Oxidation of 8b can be accomplished in the presence of MnO2 to provide 8c.
Scheme 9. Représentative synthesis of E-V-C(=O)-P-W-P-C(=O)~V-E
Ail H2N-V-C(=O}-P-W-P-C(=0)-V-E -------------!
9a 2 -A,
H2N-V-C(=O)-P-W-P-C(=O)-V-NH2 ---------9c
NH-V-C(=O)-P-W-P-C(=O)-V-E
9b
NH-V-C(=O)-P-W-P-C(=O)-V-NH
9d
Scheme 9 shows a general synthesis of an E-V-C(=O)-P-W-P-C(=O)-V-E molécule of the disclosure wherein, for illustrative purposes, E is ethylcarbonylamino. The treatment 5 of either 9a or 9c with one or two équivalents respectively of propîonyl chloride under basic conditions (e.g. sodium hydroxide) provides the molécule 9b or 9d.
Scheme 10. Représentative synthesis of R’-P-W-P-R2
Scheme 10 shows an altemate general synthesis of an R’-P-W-P-R2 intermediate of the invention wherein, for illustrative purposes, R1 and R2 are independent protecting groups and W is a two aromatic ring unit constructed via a transition métal mediated cyclization. Bromînation of 6b with a brominating agent (i.e. pyridinium tribromide) provides the dibromide 10a. Displacement of the primary bromide then proceeds by the addition of an acid under basic conditions (e.g. K2CO3) to provide lOd. Conversion to lOf or 10g can be 5 accomplished following methods described in Scheme 8.
Scheme 11. Représentative synthesis of E-V-C(=O)-P-W-P-R
O
Scheme 11 shows an altemate general synthesis of an E-V-C(=O)-P-W-P-R intermediate of the invention wherein, for illustrative purposes, Ris a protecting group and W is a two aromatic ring unit. Bromînation of 6b with a brominating agent (i.e. pyridinium tribromide) provides the dibromide 10a. Displacement of the primary bromide then proceeds by the addition of an acid under basic conditions (e.g. K2CO3) to provide 11b. Conversion to lld or lie can be accomplished following methods described in Scheme 8.
Scheme 12. Représentative synthesis of R’-V-C(=O)-P-R2
PG t
HO
V-NH-PG O
12e
Scheme 12 shows a general synthesis of an R’-V-C(=O)-P-R2 intermediate wherein, for illustrative purposes, P is pyrrolidine, R* is a generic group that is depicted as either -E or 5 a amino protecting group, and R2 is a generic group that is depicted as -C(=O)-O-PG.
Coupling of amine 12a (or 12d) with acid 12b or 12e is accomplished using a peptide coupling reagent (e.g. HATU) to afford 12c (or 12f) respectively. The conversion of 12f to 12c can be accomplished by removal of the appropriate protecting group, followed by treatment with methyl chloroformate under basic conditions (e.g. sodium hydroxide).
Scheme 13. Représentative synthesis of E-V-C(=O)-P-W-P-C(=O)-V-E
Scheme 13 shows an altemate general synthesis of an E-V-C(=O)-P-W-P-C(=O)-VE intermediate of the invention wherein, for illustrative purposes, W is a two aromatic ring unit. Displacement of the both bromides proceeds by the addition of an acid under basic conditions (e.g. K2CO3) to provide 11c. Conversion to lld or lie can be accomplished following methods described in Scheme 8.
Spécifie Embodiments
In one embodiment, provided is a compound of formula (I):
E‘*-Vl* -C(=O>PU -W1* -P,b-C(=O)-VIb-E,b (I) wherein:
W1* is
and W1* is optionally substituted with one or more halo, alkyl, haloalkyl, optionally 15 substituted aryl, optionally substituted heterocycle, or cyano;
Y’ is -O-CH2-, -CH2-O-, -0-C(=O)-, or -C(=O)-O-;
X5 is -CH2-CH2-, or -CH=CH-;
P,a and Plb are each independently:
*/vw
I
Ο— o—
HO , or<-°
V1* and V,b are each independently:
ο.
v Ψ r
Xy -Ά/ vv or
9 9 provided that at least one of Vu and Vlb is
»
E10 11 and E,b are each independently -N(H)(alkoxycarbonyl), -N(H)(cycloalkylcarbonyl), or -N(HXcycloalkyloxycarbonyl); or E,a-V,a taken together are R9’; or EIb-V,b taken together are R9b; and
R91 and R9b are each independently:
/JO λΧ) H-“V* .-Y° ° or O ;
or a pharmaceutically acceptable sait or prodrug thereof.
In one embodiment the disclosure provides a compound which has formula:
(A1)
P1b-C(=O}-V1b-E1b
E1
wherein the imidazole ring shown in formula Al, A2, A3, and A4 is optîonally substituted with one or more halo, haloalkyl, cyano, or alkyl;
or a pharmaceutically acceptable sait or prodrug thereof.
In one embodiment the disclosure provides a compound which has formula:
(A4) wherein the imidazole ring shown in formula A2 and A4 is optionally substituted with one or more halo, haloalkyl, cyano, or alkyl;
or a pharmaceutically acceptable sait or prodrug thereof.
In one embodiment P11 and Plb are each independently:
In one embodiment V1* and V,b are each independently:
provided that at least one of V1* and Vlb is
In one embodiment, provided is a compound of formula (I):
Ε'’-V'1 -C^Oj-P11 -W11 -P,b-C(=O)-V,b-Elb (I) wherein:
Wu is
and W1* is optionally substituted with one or more halo, alkyl, haloalkyl, or cyano;
Y’ is -O-CHi-, or-CHj-O-;
X5 is -CH2-CH2-, or -CH=CH-;
Pu and P,b are each independently:
V1* and Vlb are each independently:
provided that at least one of V1* and V,b is
E1* and E,b are each independently -N(H)(alkoxycarbonyl),
-N(H)(cycloalky!carbonyl), or -N(HXcyc!oalkyloxycarbonyl); or E1,-VI* taken together are
R9*; or E,b-Vlb taken together are R’b; and
R91 and R9b are each independently:
or a pharmaceutically acceptable sait or prodrug thereof.
In one embodiment, one of V1* and Vlb is:
, or , and the other of V1* and Vlb is
In one embodiment, one of V,a and Vlb is:
$ , and the other of V1* and V,b is
In one embodiment, one of V11 and V,b is:
and the other of V1* and V,b
In one embodiment, one of Vu and Vlb is:
I °x .
and the other ofV1* and VIb is
In one embodiment, one of Vu and Vlb is:
, and the other ofVu and Vlbis
In one embodiment, one of V1* and V,b is:
and the other ofV11 and V,b is
In one embodiment, one of V1* and V,b is:
and the other ofVu and V,bîs
In one embodiment, P11 and Plb are each independently:
.or
In one embodiment, one of V1* and V,bis:
In one embodiment, both of V** and Vlb are:
In one embodiment, one of V1’ and Vlb is:
In one embodiment, both of V1* and Vlb are:
In one embodiment, one of V1* and V,b is:
H , provided that bond (a) is connected to E1* or Elb and bond (b) is connected to the -C(=O)- group of formula (1) or (Al, A2, A3, or A4).
In one embodiment, one of V1* and V,b is:
H t provided that bond (a) is connected to Eu or Elb and bond (b) is connected to the
-C(=O)- group of formula (1) or (Al, A2, A3, or A4).
In one embodiment, one of Vu and V,b is:
H , provided that bond (a) is connected to E1* or E’b and bond (b) is connected to the -C(=O)- group of formula (1) or (Al, A2, A3, or A4).
In one embodiment, one of V1* and VIb is:
provided that bond (a) is connected to Eu or Elb and bond (b) is connected to the
-C(=O)- group of formula (1) or (Al, A2, A3, or A4).
In one embodiment, P’* and P1b are each independently:
In one embodiment, one of P,a and Plb is:
In one embodiment, one of P’* and P,b is:
In one embodiment, both of P1' and Plb are:
In one embodiment, -V^-C^Oy-P11- and -P,b-C(=O)-Vlb- are each independently;
provided that at least one of V1* and V,b is
In one embodîment, -^^-0(=0)^^- and -P,b-C(=O)-Vlb- are each independently:
In one embodîment, one of-V'*-C(=O)-P1*- and -P,b-C(=O)-Vlb- is:
and the other of-V’-Cf^)-?”- and -P,b-C(=O)-V,b- is:
In one embodiment, one of-V^-C^OyP1’- and -P,s-C(=O)-Vlb- is:
* and the other of-V-C(=O)-Pu- and -P,b-C(=O)-V,b- is:
In one embodiment, both of-V^-Cf^OJ-P11- and -P,b-C(=O)-Vlb- are:
In one embodiment, at least one of E1* and E,b is -N(H)(alkoxycarbonyl). 5 In one embodiment, both of E1* and EIb are -N(H)(alkoxycarbonyl).
In one embodiment, at least one of E1* and E,b is -N(H)C(=O)OMe.
In one embodiment, both of Eu and E,b are -N(H)C(=O)OMe.
In one embodiment, the disclosure provides a compound of formula:
or a pharmaceutically acceptable sait or prodrug thereof.
In another embodiment, the disclosure provides a compound of formula:
Ο /
or a pharmaceutically acceptable sait or prodrug thereof.
In one embodiment, the disclosure provides a compound of formula:
In one embodiment, the disclosure provides a compound of formula:
The disclosure will now be illustrated by the following non-limiting Examples. The following abbreviations are used throughout the spécification, încluding the Examples.
%F % Bioavaîlability
(g) Gas
°C Degree Celsius
Ac Acetate
ACN Acetonitrile
approx./apprx. Approximate
AUC Area under the curve
Bn Benzyl
BOC/Boc tert-Butoxycarbonyl
br Broad
calc'd Calcul ated
CC50 50% Cytotoxicity concentration
d Doublet
dba dibenzalacetone
DCM Dichloromethane
dd Doublet of doublets
DIPEA/DIEA N,N-Diisopropylethylamine
DMA N,N-Dimethylacetamide
DMAP 4-Dimethylaminopyridine
DMEM Eagle's minimal essential medium
DMF Dimethylformamide
DMSO/dmso Dîmethylsulfoxide
dppf l,l'-bis( diphenylphosphanyl) ferrocene
ECso Half maximal effective concentration
EDTA Ethylenediaminetetraacetic acid
ESI Electrospray ionization
Et Ethyl
FBS Fêtai bovine sérum
g Gram
HATU 2-( 1 H-7-Azabenzotriazol-l -yl)-l, 1,3,3-tetramethyl uronium hexafluorophosphate Methanaminium
HPLC High performance liquid chromatography
hr/h Hour
Hz Hertz
id. Inner diameter
IP Am Isopropylamine
IV Intravenous
J Coupling constant
L Liter
LCMS Liquid chromatography mass spectrometry
M Molar
m Multiplet
m/z Mass to charge
M+ Mass peak
Me Methyl
mg Milligram
MHz Mégahertz
min Minute
mL Milliliter
mL Milliliter
mM Millimolar
mm Millimeter
mmol Millimole
MS Mass spectrometry
MTBE Methyl tert-butyl ether
N Normal
NADPH Nicotinamide adenine dinucleotide phosphate
NB S N-Bromosucci ni m ide
nm Nanometer
NMR Nuclear magnetic résonance
o/n Over night
Papp Apparent permeability
PBS Phosphate bufier System
Pd/C Palladium on carbon
PEG Polyethylene glycol
Ph Phenyl
Piv Pivalate
Py/pyr Pyridine
q Quart et
quant Quantitative
rt/RT Room température
s Singlet
SFC Supercritical fluid chromâtography
SPhos/S-Phos 2-Dicyclohexylphosphino-2',6'-dimethoxybiphenyl
SRM Selected reaction monitoring
t Triplet
t-Bu tert-Butyl
TEA Triethylamine
TEMPO (2,2,6,6-Tetramethyl-piperidin-l-yl)oxyl
Tf Trifluoromethanesu 1 fo nate
TFA Trifluoroacetic acid
THF Tetrahydrofuran
TLC Thin layer chromatography
TM S Trimethylsilyl
UV Ultraviolet
w/w Weight to weight
X-Phos/XPhos 2-Dicyclohexylphosphino-2',4',6t ni sopropylbi pheny 1
δ Chemical shift
pL Microliter
pm Micromolar
Examples
Intermediate 1
Br 0 0
· HOrŸS - K2CO3, DMF __ Γ° V'^Tl
rt, 18 hr. ΊΠ fl
7-hydroxy-1 -tetralone 89%
1-bromo-2-(bromomethyl)- 4-chlorobenzene 7-(2-bromo-5-chlorobenzyloxy)- 3.4-dihydronaphthalen-1 (2H)-one
Pd(OPiv)2, P(4-F-Ph)3, FBuCO2H,
------------------►
K2CO3, DMA, 60 °C, 24 hr.
67-85%
3-ch!oro-10,11-dihydro-5Hdi benz o[c,g]chrom en-8 (9H)-o n e
3-chloro~10,11-di hydro-5Hdibenzo[c,gjchromen-8(9H)-one potassium virryltri fluor o borate, PdfOAch, SPhos, K2CO3 propanol (reflux)
3-vinyH0,11-dihydro-5Hcfi benzo[c,g|chromen-3(9H)-one
1. NBS HjO/THF/DMSO
2. MnOj, DCM
3-(2-bromoacetyl}-10,11 -dihydro-5Hdibenzo[c,g]chromen-8(9H)-one
7-(2-Bromo-5-chIorobenzyIoxy)-3,4~dihydronaphthalen-l(2II)-one
To a stirred solution of 7-hydroxy-l-tetralone (13.9 g, 85.7 mmol) and l-bromo-2(bromomethyl)-4-chlorobenzene (25.6 g, 90.0 mmol) in di methyl formamide (850 mL) was added potassium carbonate (24 g, 172 mmol). The reaction was stirred under argon for 18 hours then diluted with ethyl acetate (1 L). The organics were washed three times with water and once with brine. The organic layer was then dried with magnésium sulfate, filtered and concentrated. To the resulting oi! was added methanol (500 mL) and the suspension was agitated forthirty minutes. 7-(2-bromo-5-chlorobenzyloxy)-3,4-dihydronaphthalen-l(2H)one (27.8 g, 89% yield) was isolated by filtration.
3-Chloro-10,ll-dihydro-5H-dibenzo[c,g]chroinen-8(9H)-one
To a 1 L flask contaîning palladium(ll) pîvalate (1.18 g, 3.8 mmol), tri(4fluorophenyl)phosphine (1.20 g, 3.8 mmol), pivalic acid (2.33 g, 22.8 mmol) and potassium carbonate (31.8 g, 228 mmol) was added a solution of 7-(2-bromo-5-chlorobenzyloxy)-3,4dihydronaphthalen-l(2H)-one (27.8 g, 76.2 mmol) in dimethyacetamide (380 mL). The flask was evacuated and backfilled with argon 5 times and then stirred under argon at 60 °C for 24 hours. The reaction was cooled to room température and diluted with MTBE and water. The resulting biphasic mixture was stirred for 3 hours and filtered through Celite, rinsing with MTBE. The organic layer ofthe filtrate was separated and then washed twice with water and once with brine. The organics were then dried with magnésium sulfate, filtered, concentrated and purified by flash column chromatography (Hexanes/DCM) to yield 3-chloro-10,l 1dihydro-5H-dibenzo[c,g]chromen-8(9H)-one (14.4 g, 67% yield) as an off-white solid.
3-VinyH0,ll-dihydro-5H-dibenzo[c,g]chronien-8(9H)-one
A 3-neck oven-dried 500 mL round-bottom flask was cooled under Ar, then charged with 3-Chloro-10,l l-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one (12.0 g, 42.1 mmol), potassium vinyltrifluoroborate (8.47 g, 6.32 mmol), Pd(OAc)2 (473 mg, 2.11 mmol), SPhos (1.74 g, 4.25 mmol), K2CCh(17.5 g, 126 mmol) and anhydrous propano! (120 mL). The reaction mixture was sparged with Ar for 16 min, then heated to reflux for 5.5 h. Upon completion, the reaction mixture was cooled to RT and concentrated under reduced pressure. The crude residue was suspended in DCM, then washed with H2O and brine. The organic solution was dried over MgSCh, filtered and concentrated under reduced pressure. The resulting residue was further purified via silica plug, eluting with DCM to afford 3-vinyl10,ll-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one (10.2 g, 87%).
3-(2-Bromoacety 1)-10,1 l-dihydro-5H-dibenzo[c,g]chromen-8(9II)-one
3-Vinyl-10,l l-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one (9.98 g, 36.1 mmol) was dissolved in a stirred solution of THF (70 mL), DMSO (70 mL) and H2O (35 mL). NBS (6.75 g, 37.9 mmol) was added in a single portion and the reaction mixture was stirred at RT for 33 min. Upon completion, the reaction medium was diluted with EtOAc and washed twice with H2Üand once with brine. The organic phase was dried over MgSO4, filtered and concentrated under reduced pressure. The resulting crude bromohydrin was suspended in DCM (200 mL) and treated with activated MnOi (62.7 g, 722 mmol). After stirring for 15 h at RT, the reaction mixture was filtered over celite and the filter cake was rinsed several times with DCM. The combined filtrate (-400 mL) was treated with MeOH (—100 mL) and the mixture was gradually concentrated under reduced pressure, causing solid material to precipitate from solution. When the liquid volume reached -200 mL, the solid was filtered off and rinsed with MeOH. The concentration/precipitatation/filtration/rinsing sequence was performed 2x more, resulting in the collection of 3 crops of powdered 3-(2-bromoacetyl)-10,l l-dihydro-5Hdibenzo[c,g]chromen-8(9H)-one (7.49 g, 56% over 2 steps).
Intermedia te 2
P/HBrj
------>
DCWMeOH
3-(2-bromoacetyf)-10,11-dihydro5H-dibenzo[c.p)chramerve(9H}one
9-bromo-3^2-bromoacetyD'10,11dhydro-5H-<Jibenïo[c.pjéhrom»nC(9W)-one
9-Bromo-3-(2-bromoacetyl)-10,ll-dihydro-5H-<libenzo[c,g]chromen-8(9n)-one
A mixture of 3-(2-bromoacetyl)-10,l l-dihydro-5H-dîbenzo[c,g]chromen-8(9H)-one (2.58 g, 6.95 mmol), pyridinium tribromide (2.56 g, 8.0 mmol), dichloromethane (22mL) and methanol (2.5mL) was stirred at about 20°C for 3 hours to obtain a slurry. The precipitated product was filtered, washed with dichloromethane (lOmL) and dried in a vacuum oven at 40°C to give 9-bromo-3-(2-bromoacetyl)-10,l l-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one (2.62 g, 84% yield). 400 MHz Ή NMR (CDCb) δ 8.03-8.01 (m, 1H), 7.85 (d, J= 8.2 Hz, 1H), 7.82 (s, 1H), 7.71 (s, 1H), 7.67 (s, 1H), 5.19 (s, 2H), 4.74 (dd, J = 4.1,4.1 Hz, 1H), 4.45
(s, 2H), 3.37-3.29 (m, 1H), 2.99-2.92 (m,lH), 2.59-2.46 (m, 2H).
Intermediate 2a
—Q _0 * °
Η<λ /=\ /P y=\ ,P
Br—' \_y-Br -------► Br—' \=^_J>-Br
3-P-bromo-1-hydroxyethyt)-10,11- &*romt>3-(2-bromo-1dihydro-5H-dibenzo(c jjchromen- h/<io>tyetryt}-1011-dr>ydro-5H· 9-bfOmo-3-p^nxnoacety1}-10.116(9 H)-one dibenzo(c,o)chroner>8(9H)-one <ihydro-5H-<*benzo(c,i)ctTOmen8(9H}-one
9-Bromo-3-(2-bromoacetyl)-10,ll-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one
To 3-(2-bromo-l-hydroxyethyl)-10,11 -dihydro-5H-dîbenzo[c,g]chromen-8(9H)-one (20.3g, 54.4 mmol) in DCM (365 mL) was added MeOH (22 mL) and pyridinium tribromide (18.24 g, 57.0 mmol). After 2h, water was added (lOOmL) and after briefly agitating the layers split and the bottom organic layer was collected. The organic layer was then washed with IM HCl (100 mL) and the bottom organic layer containing 9-bromo-3-(2-bromo-lhydroxyethyl)-10,l l-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one was collected. 400 MHz ’H NMR (CDCb) 7.75 (d, J = 8.1 Hz, 1H), 7.68 (s, 1H), 7.61 (s, 1H), 7.42 (d, J = 7.5 Hz, 1H), 7.24 (s, 1H), 5.13 (s, 2H), 4.99-4.96 (m, 1H), 4.73 (dd, J = 4.1, 4.1 Hz, 1H), 3.69-3.66 (m, 1H), 3.58-3.53 (m, 1H), 3.35-3.27 (m, 1H), 2.96-2.90 (m, 1H), 2.58-2.44 (m, 2H), C-OH not observed.
To 9-bromo-3-(2-bromo-l-hydroxyethyl)-l 0,1 l-dihydro-5H-dibenzo[c,g]chromen8(9H)-one (approx. 54.4 mmol) in DCM (365mL) was added sodium bicarbonate (5.45 g), sodium bromide (6.14 g), TEMPO (16.55 mg) and water (60 mL). The solution was cooled between 0-5 °C and 6% bleach (91.5 mL) was added. After Ih isopropyl alcohol (20 mL) was added and the reaction mixture was warmed to room température. Agitation was stopped, the layers separated and the lower organic layer was collected and concentrated removing approximately 345 g of solvent. The slurry was filtered and the cake washed with 50 mL water and then 50 mL DCM (pre-cooled to 5 °C). The solids were collected and dried under vacuum to obtain 9-bromo-3-(2-bromoacetyl)-10,ll-dihydro-5H-dibenzo[c,g]chromen8(9H)-one(18.6g, 76% yield). 400 MHz *H NMR (CDCb) Ô 8.03-8.01 (m, 1H), 7.85 (d, J= 8.2 Hz, 1H), 7.82 (s, 1H), 7.71 (s, 1H), 7.67 (s, 1H), 5.19 (s, 2H),4.74(dd, J= 4.1,4.1 Hz, 1H), 4.45 (s, 2H), 3.37-3.29 (m, 1H), 2.99-2.92 (m,lH), 2.59-2.46 (m, 2H); 100 MHz nC NMR(CDCh) δ 190.4, 189.6, 154.2, 136.6, 134.1, 133.9, 132.9, 131.8, 129.3,127.2, 125.6, 124.2, 123.3, 117.0, 68.1,49.9,31.8,30.4,25.5.
Intermediate 2b Cls^h » PdCyMeCNfe, X-Phos K3PO4, MeCN. 65’C
3^(trimethyfsi>yl)ethynyf)-10,11-dihydro5H-dfoenzoIc,g|chromen-â{9H)-one
3-ditoro-10,11-dihydrt>-5Hdibenzo(c,p]chromenre(9H>or>e
3-acetyM 0,11 -dihydro-5Hdibenzo[c^chromerH8{9H)-one
PyHBr3
DCM, MeOH *C
9-bromo-3-(2-bromoacetyl)-10.11-dihydro-5Hdtoenzo[c, chrome n-B(9H)-one
3-((TrimethylsiIyl)ethynyl)-10,ll-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one
A 300 mL flask equipped with an overhead stirrer and a reflux condenser under an atmosphère of nitrogen was charged with 3-chIoro-10,l l-dihydro-5H-dibenzo[c,g]chromen8(9H)-one (10.0g, 35.12 mmol), powdered anhydrous tripotassium phosphate (22.4 g, 105.4 mmol), XPhos (1.34 g, 2.81 mmol), and PdCh(MeCN)2 (364 mg, 1.40 mmol). Acetonitrile (140 mL) was added followed by TMSacetylene (18 mL, 141 mmol). The mixture was heated to 65 °C. After 6h, the reaction was judged complété, and the mixture was cooled to 20 °C. The mixture was filtered through a fritted funnel, and the filtercake was washed with acetonitrile. The filtrate was concentrated to about 150 mL under reduced pressure and extracted with heptane (50 mL, 3*100 mL). jV-Acetyl cysteine (15 g) was added to the acetonitrile phase, and the mixture was agitated for 5 h at 45 °C. The mixture was cooled to ambient température, filtered through a fritted fùnnel, and the filtercake was washed with acetonitrile. The filtrate was concentrated to about 120 mL under reduced pressure. Water (120 mL) was added and the mixture was agitated for 40 minutes at 45 °C and then cooled to ambient température. After 30 minutes the mixture was filtered through a fritted fùnnel to provide 3-((trimethylsilyI)ethynyl)-10,l l-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one (4.07 g, 33.4% yield ) as a yellow solid: 400 MHz *H NMR (CDCb) δ 7.65 (d, J = S.l Hz, 1H), 7.60 (s, 1H), 7.55 (s, 1H), 7.47 (dd, J = 8.1,1.4 Hz, 1 H), 7.27 (s, 1H), 5.06 (s, 2H), 2.95 (ζ J = 6.1 Hz, 2H), 2.67 - 2.59 (m, 2H), 2.18 - 2.08 (m, 2H), 0.26 (s, 9H).
3-Acetyl-10,ll-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one
A 20 mL vial with stirbar was charged with 3-((trimethy 1 si lyl)ethyny 1)-10,11-dihydro5H-dibenzo[c,g]chromen-8(9H)-one (850 mg, 2.44 mmol) and formic acid (9.8 mL). The solution was heated to 65 °C. After 3 h, the reaction was judged complété. The mixture was concentrated under reduced pressure; the resulting residue was taken up in CH2CI2 and loaded onto a prepacked 25g silica gel cartridge. The product was purified by chromatography on a prepacked 80g silica gel column eluting with a solvent gradient from 5% to 85% EtOAc/hexanes. The product containing fractions were combined and concentrated to provide 3-acety!-10,ll-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one (616 mg, 86%): 400 MHz Ή NMR (CDCb) δ 8.00 - 7.94 (m, 1H), 7.81 (d, J = 8.2 Hz, 1H), 7.77 (s, 1H), 7.64 (s, 2H), 5.16 (s, 2H), 2.98 (t, J = 6.1 Hz, 2H), 2.69 - 2.64 (m, 2H), 2.63 (s, 3H), 2.21-2.09 (m, 2H)
9-Bromo-3-(2-bromoacetyl)-10,l l-dihydro-5II-dibenzo|c,g|chromen-8(9H)-one
A 20 mL vial with a stirbar was charged with 3-acetyl-10,l l-dihydro-5Hdibenzo[c,g]chromen-8(9H)-one (100 mg, 0.366 mmol), 9:1 CHiClî/MeOH (3.4 mL) and pyridinium tribromide (246 mg, 0.769 mmol). The solution was heated to 35 °C. After 30 minutes, the reaction was judged complété. The mixture was cooled to ambient température, diluted with EtOAc (50 mL) and sequentially washed with saturated aqueous NaîSîOî (20 mL), 2% aqueous NaHCOj (20 mL), water (20 mL), and brine (10 mL). The organic phase was dried over MgSOi, filtered and concentrated under reduced pressure resulting in 9bromo-3-(2-bromoacetyl)-10,l l-dihydro-5H-dibenzo[c,g]chromen-8(9H)-one (68 mg, 41%): 400 MHz Ή NMR (CDCb) δ 8.03 - 8.01 (m, 1H), 7.85 (d, J= 8.2 Hz, 1H), 7.82 (s, 1H), 7.71 (s, 1H), 7.67 (s, 1H), 5.19 (s, 2H), 4.74 (dd,J=4.1,4.1 Hz, 1H),4.45 (s, 2H), 3.37-3.29 (m, 1H), 2.99 - 2.92 (m,lH), 2.59 - 2.46 (m, 2H).
CHrMgBr
Intermediate 3
THF, -40-0 °C
TFA
DCM
H2(g)
Pd/C (S)-ethyl 2-(tertbutoxycartxjnylamino)-i>oxohexanoate
(2S,5S)-ettiyl 5methylpyrrolidine-2carboxylate
DIEA, DMAP (Boc)2O
DCM (2S,5S)-1-tert-butyl 2-ethyl 5methylpyrrolidine-1,2dicarboxylate
UOH
EtOH. H2O
0-CH3
O Boc (2S,5S)-1-(tertbutoxycarbonyl)-5· methylpyrrolidine2-carboxylic acid (S)-Ethyl 2-(tert-biitoxycarbonylammo)-5-oxohexanoate.
A solution of ethyl jV-Boc (S)-pyroglutamate (20.0 g, 77.7 mmol) was in anhydrous
THF (150 mL) in a two neck round bottom under argon was cooled to -40 °C. Méthylmagnésium bromide solution (3.0 M in Ether, 28.5 mL, 85.5 mmol) was added to the reaction mixture dropwise over 30 minutes. The reaction was stirred for 4 hrs at -40 °C then for 1 hr at 0 °C. The reaction was partitioned between ethyl acetate and saturated ammonium chloride solution and acidified with 1 N HCl. The aqueous layer was extracted two more times with ethylacetate. The organic layers were combined and dried with sodium sulfate. The crude materiel was purified by column chromatography (20% - 40% EtOAc/hexanes) to yield (S)-ethyl 2-(tert-butoxycarbonylamino)-5-oxohexanoate as a viscous oi! and was used directly in the following step.
(S)-Ethyl5-m ethyI-3,4-d ihy d ro-2II-py rroIe-2-ca rbo xy late.
(S)-ethyl 2-(tert-butoxycarbonylamino)-5-oxohexanoate in a 1 L flask was treated with a trifluoro acetic acid / dichloromethane solution (1:1 mixture, 100 mL). Effervescence was observed and the mixture was allowed to stir for 4 hours at room température. After which time the volatiles were removed in vacuo to yield (S)-ethyl 5-methyl-3,4-dihydro-2Hpyrrole-2-carboxylate as an oil, and used directly in the following step.
(2S,5S)-Ethyl 5-methylpyrroIidine-2-carboxylate.
(S)-Ethyl 5-methyl-3,4-dihydro-2H-pyrrole-2-carboxylate in a IL flask was dissolved with éthanol (400 mL) was evacuated and charged with argon three times (3x). Palladium on carbon (apprx. 750 mg, 10% w/w, dry) was added and the reaction was evacuated of gas and charged with hydrogen gas (3x). The reaction was allowed to stir under atmospheric hydrogen for 16 hours. The mixture was filtered through a plug of celite and the filtrate was concentrated in vacuo. Diethyl ether was added to the oil and a precipitate formed. The mixture was filtered to yield (2S,5S)-ethyl 5-methylpyrrolidine-2-carboxylate, as a white solid (10.6 g, 67.4 mmol, 86.7% over three steps). ’H NMR (400 MHz, cdcb) δ 4.48 (dd, 1H), 4.27 (q, 2H), 3.92-3.80 (m, 1H), 2.52 - 2.36 (m, 1H), 2.32-2.13 (m, 2H), 1.75 - 1.60 (m, 1H), 1.51 (d, 3H), 1.30 (t, 3H).
(2S»5S)-l-Tert-butyl 2-ethyl 5-methylpyrrolidine-l,2-dicarboxylate.
To a solution of (2S,5S)-ethyl 5-methylpyrrolidine-2-carboxylate (7.0 g, 44.5 mmol) in dichloromethane (250 mL), ditertbutylanhydride (10.7 g, 49.0 mmol), diisopropylethylamine (17.1 mL, 98.0 mmol) dropwise over 10 minutes, and dimethyl amino pyridine (0.27 g, 2.23 mmol) were added successively. Effervescence was observed and the mixture was allowed to stir for 16 hours at room température. The reaction was washed with HCl (250 mL, of IN). The organic layer was then dried with sodium sulfate. The crude material was purified by column chromatography (5% - 25% EtOAc/hexanes) to yield (2S,5S)-l-tert-butyl 2-ethyl 5-methylpyrrolidine-l,2-dicarboxylate as an oil (6.46 g, 25.1 mmol, 56%). LCMS-ESI*: calc’d for C13H23NO4: 257.16 (M +); Found: 258.70 (M+H+).
(2S,5S)-l-(Tert-butoxycarbonyl)-5-methylpyrrolidine-2-carboxylÎc acid.
To a solution of (2S,5S)-l-tert-butyJ 2-ethyl 5-methylpyrTo!idine-l,2-dicarboxylate (6.46 g, 25.1 mmol) in éthanol (20 mL) was added lithium hydroxide mono hydrate (2.11 g, 50.2 mmol) and deionized water (12mL). The mixture was allowed to stir for 16 hours then partitioned between ethylacetate and a 1:1 mixture of saturated brine and IN HCl. The aqueous layer was extracted an additional time with ethyl acetate. The organic layers were combined, dried with sodium sulfate and the solvent was removed in vacuo to yield (2S,5S)l-(tert-butoxycarbonyl)-5-methy!pyrrolidine-2-carboxy!ic acid as a white solid (quant.) and was used directly in the following step.
Intermediate 4
O HOA^NHCO2Me
HATU, DIPEA DMF
TFA
(2S,5S)-ethyl 5methylpyrrolidine-2carboxylate-TFA (S)-2(methoxycarbonylamino) -3-methylbutanoic acid
NHCO2Me °rV
Etc)Cj
LiOH H2O/MeOH >NHCO2Me °rV
VS(2S,5S)-ethyl 1-((S>-2(methoxycarbony!amino)-3methylbutanoyl)-5methylpyrrolidine-2-carboxylate (2S,5S)-1-((S)-2(methoxycartxjnyIamino)-3methylbutanoyl)-5methylpyrrolidine-2-carboxylic acid (2S,5S)-EtliyI l-((S)-2-(methoxycarbonyIamino)~3-ntethylbutanoyl)-5~ methylpyrrolidine-2-carboxylate.
(2S,5S)-Ethyl 5-methylpyrrolidîne-2-carboxylate 2,2,2-trifluoroacetate (10.0 g, 39.3 mmol), (S)-2-(methoxycarbonylamino)-3-methylbutanoic acid (6.88 g, 39.3 mmol) and HATU (14.9 g, 39.3 mmol) were combined in DMF (100 mL) and DIPEA (15.0 mL, 86.5 mmol) was added. After stirring for 1 h at RT, the reaction mixture was diluted with EtOAc. The organic phase was washed successively with 10% HCl, saturated aqueous NaHCO3 and brine, then dried over MgSO4, filtered and concentrated under reduced pressure to afford (2S,5S)-ethyl l-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)-5-methylpyrrolidine-2carboxylate. The crude material was carried on without further purification.
(2S,SS)-l-((S)-2-(Methoxycarbonylamino)-3-methylbutanoyl)-5-methylpyrrolidine-2carboxylic acid.
(2S,5S)-Ethyl l-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)-5methylpyrrolidine-2-carboxylate (39.3 mmol, assuming complété conversion from the previous transformation) was suspended in MeOH (200 mL) and aqueous LiOH (1.0 M, 100 mL, 100 mmol) was added. The reaction mixture was stirred o/n, then concentrated under reduced pressure to remove most of the MeOH, The aqueous solution was washed 2x with DCM before being acidified to pH-1-2 with 10% HCl. The acidic aqueous phase was then extracted 5x with EtOAc. The combined EtOAc extracts were dried over MgSO4 filtered and concentrated under reduced pressure to afford (2S,5S)-l-((S)-2-(Methoxycarbonylamino)-35 methylbutanoyl)-5-methylpyrrolidine-2-carboxylic acid (6.89 g, 56% over 2 steps).
Intermediate 5
ÇN h47> MeO2C (2S,4S)-1-ferf-butyl 2-methyl 4-cyanopyrrolidine-12~ dicarboxylate
1) HCl. MeOH
2) BocjO, NaHCOj
ÇO2Me
NaOH Boc (2S,4S)-1-fert-butyl 2,4dimethyl pyrrolidine-1,2,4tricarboxylate
CO2H
1) EtO2CCI
OS,5 S)-1 -(terf-butoxyca rbonyf)-5(methoxycarbonyl)pyrrolidine-3carboxylic acid
Me! (t-Bu)2pyr
AgOTf
1-(tert-butyl) 2-methyl (2S.4SH(hydroxymethyf)pyrrolidine-1,2dicarboxylate
LiOH
1-(terf-butyl) 2-methyl (2S,4S)-4(methoxymethyl)pyrrolidine-l dicarboxylate (2S,4S)-1 -<fert-butoxycarbonyl)-4(methoxymethyl)pyrrolîdine-2carboxylic acid (2S,4S)~l~Tert-biityl 2,4-dimethyl pyrro!idine-l,2,4-tricarboiylate.
To a solution of (2S,4S)-l-tert-butyl 2-methyl 4-cyanopyrrolidine-l,2-dicarboxylate (9.0 g, 35.4 mmol) in MeOH (196 mL) was added HCl (4M in 1,4-dioxane, 100 mL, 403 mmol). The solution was stirred at room température for 16h and concentrated in vacuo. The crude intermediate was dissolved in EtOAc (180 mL) and basified with aqueous bicarbonate (sat.). Di-tert-butyl dicarbonate (8.5 g, 38.9 mmol) was added and the biphasic solution was stirred at room température for 12h. The layers were then separated and the aqueous layer was back extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, and concentrated. The crude oil was purified by silica gel chromatography (15% to 40% to 100% EtOAc/Hexanes) to provide (2S,4S)-l-tert-buty! 2,4-dimethyl pyrTolidine-l,2,4-tricarboxylate (9.56 g, 94%).
(3S^S)-l-(Tert-butoxycarbonyl)-5-(methoxycarbonyl)pyrrolidine-3-carboxylic acid.
To a solution of (2S,4S)-l-tert-butyl 2,4-dimethyl pyrrolidine-l,2,4-tricarboxylate (9.56 g, 33.3 mmol) in THF (70 mL) at 0 °C (extemal température, ice bath) was added NaOH (IN aqueous, 33 mL, 33.3 mmol) dropwise over 15 min. The solution was stirred at 0 °C for 5h before acidification with HCl (IN). The solution was extracted with EtOAc (3x). The combined organic layers were dried over Na2SO4 and concentrated. The crude oil was purified by silica gel chromatography (2% to 5% to 10% MeOH/CH2Cl2) to provide (3S,5S)l-(tert-butoxycarbonyl)-5-(methoxycarbonyl)pyrroÎidine-3-carboxylic acid (6.38g, 70%).
(25.45) -l-Tert-butyl 2-methyl 4-(hydroxymethyl)pyrrolidine-l,2-dicarboxylate.
To a solution of(3S,5S)-l-(tert-butoxycarbonyl)-5-(methoxycarbonyl)pynOlidine-3carboxylic acid (6.38 g, 23.3 mmol) in THF (116 mL) at 0 °C (extemal température, ice bath) was added Et3N (4.9 mL, 35.0 mmol) and ethyl chloroformate (2.7 mL, 28.0 mmol). The resulting solution was stirred at 0 °C for 45 min, during which time a white precipitate forms. The reaction mixture was filtered through celite and concentrated.
The crude intermediate was dissolved in THF (59 mL) and cooled to 0 °C (extemal température, ice bath). NaBHi (4.41 g, 116.7 mmol) in H2O (59 mL) was slowly added and the resulting solution was stirred at 0 °C for 2 h. The reaction mixture was diluted with EtOAc and washed with H2O. The aqueous layer was back extracted with EtOAc. The combined organic layers were dried over Na2SÛ4 and concentrated. The crude oil was purified by silica gel chromatography (42% to 69% to 100% EtOAc/Hexanes) to provide (2S,4S)-l-tert-butyl 2-methyl 4-(hydroxymethyl)pyrrolidine-l,2-dicarboxylate (3.63 g, 60%).
(25.45) -l-Tert-buty! 2-methyl 4-(methoxymethyI)pyrrolidine-l ,2-dicarboxylate.
To a solution of (2S,4S)-l-tert-butyl 2-methyl 4-(hydroxymethyl)pyrrolidine-l,2dicarboxylate (2.57 g, 9.9 mmol) in CH2CI2 (50 mL) was added AgOTf (4.07 g, 15.8 mmol) and 2,6-di-/er/-butylpyridine (4.4 mL, 19.8 mmol). The reaction mixture was cooled to 0 °C (extemal température, ice bath) and Mel (0.98 mL, 15.8 mmol) was slowly added. The resulting slurry was stirred at 0 °C for 1.5 h and at room température for 1.5 h. The slurry was diluted with CH2C12 and filtered through celite. The filtrate was concentrated to dryness, dissolved in Et2O, and washed with HCl (IN) and brine. The aqueous layers were backextracted with EtîO and the combined organic layers were dried over NaîSCh and concentrated. The crude oil was purified by silica gel chromatography (10% to 75% to 100% EtOAc/Hexanes) to provide (2S,4S)-l-teTt-butyl 2-methyl 4-(methoxymethyl)pyrroIidine-l,2dicarboxylate (2.11 g, 78%). *H-NMR: 400 MHz, (CDCh) δ: (mixture of rotomers, major reported) 4.20 (t, 1H), 3.71 (s, 3H), 3.67 (m, 1H), 3.34 (m, 2H), 3.30 (s, 3H), 3.16 (t, 1H), 2.43 (m, 2H), 1.74 (m, 1H), 1.38 (s, 9H).
(2S,4S)-l-(Tert-butoxycarbonyl)-4-(methoxymethyl)pyrrolidine-2-carboxylic acid.
To a solution of (2S,4S)-l-teTt-butyl 2-methyl 4-(methoxymethyI)pyrroIidine-l,2dicarboxylate (2.11 g, 7.7 mmol) in a mixture of THF (38 mL) and MeOH (15 mL) was added LiOH (2.5 M aqueous, 15 mL, 38.6 mmol). The resulting solution was stirred at room température for 2h, and acidified with aqueous HCl (IN). The desired product was extracted with CH2CI2 (4x). The combined organic layers were dried over Na2SO4 and concentrated to provide (2S,4S)-l-(tert-butoxycarbonyl)-4-(methoxymethyl)pynOlidine-2-caiboxylic acid (2.0 g, 99%). ’H-NMR: 400 MHz, (CDCb) δ: (mixture of rotomers, major reported) 4.33 (t, 1H), 3.65 (m, 1H), 3.35 (m, 2H), 3.32 (s, 3H), 3.16 (t, 1H), 2.45 (m, 2H), 2.12 (m, 1H), 1.46 (s,9H).
Intermediate 6
(2S,5S)-l-((2S3S)-2-(Methoxycarbonylamino)-3-methyIpentanoyl)-5 methylpyrrolidine2-carboxylic acid.
(2S,5S)-1-((2S,3 S)-2-(methoxy carbonyl amino)-3 -methy Ipentanoyl)-5 methylpyrrolidine-2-caiboxyIic acid was synthesized in a similar manner as Intermediate 4 substituting (S)-2-(methoxycarbonylamino)-3-methyibutanoic acid with (2S,3S)-2(methoxycarbonyl-amino)-3-methylpentanoic acid MS (ES1) m!z 301.19 [M + H]+.
Intermediate 7
(2S,5S)-l-(Tert-butoxycarbonyl)-5-ethyIpyrrolidine-2-carboiylic acid.
(2S,5S)-l-(tert-butoxycarbonyl)-5-ethylpyrrolidine-2-carboxylic acid was synthesized in a similar manner as Intermediate 3 substituting ethylmagnésium bromide for méthylmagnésium bromide. ’HNMR (400 MHz, DMSO-d6): δ 12.37 (1H, s), 4.05-4.07 (1H, m), 3.63-3.64 (1H, m), 2.13-2.15 (1 H, m), 1.63-1.90 (4H, m), 1.39 (10H, m), 0.83 (3H, t, J = 7.2 Hz).
Intermediate 8
(2S,5S)-5-Ethyl-l-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)pyrroIidine-2carboxylic acid.
(2S,5S)-5-ethyl-l-((S)-2-(methoxycarbonylamino)-3-Tnethylbutanoyl)pyrrolidine-2· carboxylic acid was synthesized in a similar manner as Intermediate 4 substituting (2S.5S)15 ethyl 5-methylpyrrolidine-2-carboxylate 2,2,2-trifluoroacetate with (2S,5S)-methyl 5ethylpyrrolidine-2-carboxylate-HCl. MS (ESI) m!z 301.15 [M + H]+.
Intermediate 9
(2S,5S)-5-Ethyl-l-((2S3S)-2-(methoxycarbonyIamino)-3-methyIpentanoyl)pyrrolidine2-carboxylic acid.
(2S, 5 S)-5-ethyl-1 -((2 S,3 S)-2-(methoxy carbonyl amino)-3 methylpentanoyl)pyrroIidine-2-carboxyIic acid was synthesized in a similar manner as
Intermediate 4 substituting (S)-2~(methoxycarbonylamino)-3-methyIbutanoic acid with (2S,3S)-2-(methoxycarbonyl- amino)-3-methyl pentanoic acid and (2S,5S)-ethyl 5methylpyrrolidine-2-carboxylate 2,2,2-trifluoroacetate with (2S,5S)-methyl 5ethylpyrrolidine-2-carboxylate hydrochloride.
Intermediate 10
CrQj, Pyridine
DCM, rt, 4h
Ph3PEtBr
MeO 1 O^O KO’Bu, THF, rt. MeO i 0^0
4h 4-
(2S,4R)-1-tert-butyl 2-methyl 4hydroxypyrrolidine1,2-dicarboxylate (S)-1-terf-b irtyl 2-methyl 4oxopyrrolidine-1,2-dicarboxylate (S)-1-tert-butyl 2-methyl 4ethylidenepyrrolidine-1,2-dicarboxylete
10%Pd/C
H2, EtOH, rt, ovemight
(JOH
MeOH, H2O rt,2h
PhCH2Br
TEA, THF, O°C-rt ovemight (2S)-1-fert-butyl 2-methyl 4ethylpyrrolidine-1,2-dicarboxylate (2S)-1-(tert-butoxy carbonyl )-4ethylpyrrolidme-2-carboxylic add
(2S)-2-benzyl 1-tert-butyl4ethylpyrrolidtne-1,2dicarboxylate (2S,4S)-2-benzyl 1-tert-butyl 4ethylpyrrolidtne-1,2-dicarboxylate (2S,4S)-1-{tert-biJtoxycarbonyl)-4ethylpyrro!idine-2-carboxyhc acid (S)-l-Tert-butyl 2-methyl 4-oiopyrrolidine-l^-dicarboxylate
CrÜ3 (194 g, 1.94 mol) was added slowly with stirring over 30 min to a solution of pyridine (340 mL) in DCM (900 mL) at 0°C. The mixture was warmed to rt and (2S,4R)-1tert-butyl 2-methyl 4-hydroxypyrrolidine-l,2-dicarboxylate (56 g, 0.216 mol) in DCM (700 mL) was added. The réaction was stirred vigorously for 4hs at rt. The formed dark solid was decanted and washed with DCM. The organic phases were washed with aq. NaHCCh, 10% aqueous critic acid, and brine, and dried over anhydrous NaîSO4. The solvent was removed in vacuo and purified by silica gel column chromatography (PE: EtOAc=50:l to 10:1) to afford (S)-l-tert-butyl 2-methyl 4-oxopyrro!idine-l,2-dicarboxylate (42.6 g, 81%) as yellow oil.
(S)-l-Tert-butyl 2-methyl 4-ethylidenepyrrolidine-l^-dicarboxylate
A solution of PfoPEtBr (84 g, 227 mmol) and KOtBu (76.7 g, 556 mmol) in THF (1100 mL) was stirred at rt under nitrogen atmosphère for lh, and then added (S)-l -tert-butyl 2-methyl 4-oxopyrrolidine-l,2-dicarboxylate (50 g, 206 mmol) in THF (350 mL) dropwise. The mixture was stirred at room température for 4hs. TLC showed the reaction was completed. The mixture was quenched with NH4C1 aqueous and concentrated to remove THF, and then dissolved in EtOAc and water. The combined organic layer was washed with water, brine, dried over Na2SO4, filtered and concentrated. The crude product was purified by column chromatography (PE: EtOAc=30:l to 5:1) to afford (S)-1-tert-butyl 2-methyl 4ethylidenepyrrolidine-l,2-dicarboxylate (18.3 g, 35%) as yellow oil.
(2S)-l-Tert-buty! 2-methyl 4-ethylpyrrolidine-l,2-dicarboxyIate
A mixture of (S)-1-tert-butyl 2-methyl 4-ethylidcncpyrrolidinc-l,2-dicarboxylatc (50 g, 196 mmol), Pd/C (5 g) in EtOH (500 mL) was hydrogenated at room température ovemight. The mixture was filtered and concentrated to afford (2S)-1-tert-butyl 2-methyl 4-ethylpyrrolidine1,2-dicarboxylate (9.8 g, 97%) as colorless oil.
(2S)-l-(Tert-butoxycarbonyI)-4-ethylpyrrolidine-2-carboxyIic acid
A mixture of (2S)-1-tert-butyl 2-methyl 4-ethylpyrrolidine-l,2-dicarboxylate (49.5 g, 0.19 mol), LiOH (950 mL, IM) in MeOH (1500 mL) was stirred at room température ovemight. TLC showed the reaction was completed. The mixture was concentrated, adjusted the pH to 2 with IN HCl. The mixture was extracted with EA, the combined organic layer was washed with brine, dried over NaîSOi, concentrated to afford (2S)-1 -(tertbutoxycarbonyl)-4-ethytpynrolidine-2-carboxylic acid (45.5 g, 97%) as white solid without further purification.
(2S)-2-Benzyl 1-tert-butyl 4-ethyIpyrrolidine-l,2-dicarboxylate
A mixture of(2S)-l-(tert-butoxycarbonyl)-4-ethylpyrrolidine-2-carboxylÎc acid (45.5 g, 187 mmol), TEA(37.8 g, 374 mmol) in THF (1 L) was added dropwise BnBr (38.5 g, 225 mmol) at O °C. The mixture was stirred at room température ovemight. TLC showed the reaction was completed. The mixture was concentrated to remove solvent. The residue was partitioned between EtOAc and water. The combined organîc layer was washed with brine, dried over Na2SO4 and concentrated. The crude product was purified by column chromatography to give (2S)-2-benzyl 1-tert-butyl 4-ethylpyrrolidine-l,2-dicarboxylate (46 g, 74 %) as colorless oil. (2S)-2-benzyI 1-tert-butyl 4-ethylpyrrolidine-l,2-dicarboxylate was separated by préparative SFC via a Chiralcel OD 250*50mm i.d. 10 pm column (Mobile phase: A for n-hexane and B for éthanol (0.05%IPAm), Gradient: A: B = 97:3, Flow rate: lOOml/min, Wavelength: 210 and 220nm, Injection amount: 0.4g per injection) to provide (2S,4S)-2-benzyl 1-tert-butyl 4-ethy1pyrrolidine-l,2-dicarboxylate.
(2S,4S)-l-(Tert-butoxycarbonyl)-4-ethylpynOlidine-2-carboiylic acid
A mixture of (2S,4S)-2-benzyl 1-tert-butyl 4-ethy!pyrrolidine-l,2-dicarboxylate (18 g, 54.1 mmol), Pd/C (3.6 g) in MeOH (1 L) was hydrogenated at room température ovemight. TLC showed that the reaction was completed. The mixture was filtered by Celite. The filtrate was concentrated to afîord (2S,4S)-l-(tert-butoxycaibonyl)-4-ethylpyrrolidine-2-carboxylic acid (10 g, 77 %) as white solid. lH NMR: 400 MHz CDCh:ô 9.88 (br, 1H), 4.31-4.19 (m, 1H), 3.82-3.68 (m, 1H), 3.03-2.95 (m, 1H), 2.49-2.39 (m, 1H), 2.12-2.03 (m, 1H), 1.81-1.56
(m, 1H), 1.45 (d, J=8 Hz, UH), 0.92 (t, J =6 Hz, 3H).
Intermediate 11
/U EtO2CZ^g0C 1)Pt/C, h2 -----------►- 2) LiOH ..... (+/-) HOzC Boc
1-terf-butyl 2-ethyl 4,5-dimethyl-1H- rel-(2S,4S,5S)-1-(fert-butoxycarbonyl)pyrrole-1,2-dicarboxylate 4,5-dimethylpyrrolidine-2-cartx)xylic acid rel-(2S,4S,5S)-l-(tert-butoxycarbonyI)-4,5-dimethylpyrrolidine-2-carboxylic acid.
To a solution of 1-tert-butyl 2-ethyl 4,5-dimethyl-lH-pyrro!e-l,2-dicarboxylate (4.016 g, 15.02 mmol) in EtOH (100 mL) was added Platinum on carbon (5%, 0.58 g). The slurry was stirred under an atmosphère of hydrogen (1 atm) for 3 dyas. The slurry was filtered through celite and washed with MeOH. The filtrate was concentrated and the crude was purified by column chromatography (SiCh, 5-10-20% EtOAc/Hexanes) to provide rel (2S,4S,5S)-1-tert-butyl 2-ethyl 4,5-dimethylpyrrolidine-l,2-dicarboxylate.
To a solution of re!-(2S,4S,5 S)-1-tert-butyl 2-ethyl 4,5-dimethylpyrrolidine-l,2dicarboxylate in a mixture of THF (70 mL), MeOH (25 mL), and H2O (25 mL) was added lithium hydroxide (1.53 g, 63.7 mmol). The slurry was stirred at room température for 2.5h and at 45 °C for 2 h. The solution was cooled to room température and HCl (aqueous, IN, 70 mL) was added. The organics were concentrated and the resulting aqueous layer was extracted with EtOAc (3x). The combined organic layers were dried over NaîSO-i and concentrated to provide rel-(2S,4S,5S)-l-{tert-butoxycarbonyl)-4,5-dimethylpyrro!idine-2carboxylic acid (3.08 g, 84%).
Intermediate 12
IîocjO
-------->
DIPEA.DMAP
DCM
(2S3aS .GaSJ-bcnzj t octahydrocyclopenta[b]pyrrole-2carboxylate hydrochloride (2S3aS.SaS)-2-bcn7y! 1-tert-butyl hexahydrocyclopenta[t>]pynole1,2(211)-dicarboxylate
Pd/C, II2(cxccss) -----------------------------------------------------!
EtOAc
(2S3aS,6aS>1-(tertbutoxyca rbony ] )octahyd rocyclopcnt a[b]pyrrolc-2-carboxylic acid (2S,3aS,6aS)-2-benzyI 1-tert-butyl hexahydrocyclopenta[b]pyrrole-l,2(2H)dicarboxylate
To a solution of commercially available (2S,3aS,6aS)-benzyl octahydrocyclopenta[b]pyrrole-2-carboxylate hydrochloride (4.70 g, 16.68 mmol) in methylene chloride (42 mL) was added Di-tert-butyl dicarbonate (7.28 g, 33.36 mmol) N,Ndiisopropylethylamine (5.82 mL, 33.36 mmol) and 4-(Dimethylamino)pyridine (0.20 g, 1.67 mmol). The solution was stirred under air for 16 hours. Upon completion, the reaction was concentrated in vacuo, diluted in ethyl acetate, and washed with IN HCl. The aqueous layers were backextracted twice with ethyl acetate and the combined organic layers were dried over sodium sulfate, filtered and concentrated. The resulting residue was purified by sîlica gel chromatrography (5-40% ethyl acetate in hexanes) to afford (2S,3aS,6aS)-2-benzyl 1-tert butyl hexahydrocyclopenta[b]pyrrole-l,2(2H)-dicarboxylate which was used without further purification. MS (ESI) m!z 368 47 [M + Na]+.
(2S,3aS,6aS)-l-(tert-butoxycarbonyI)octahydrocyclopenta[b]pyrrole-2-carboxylic acid
To a 250mL round bottom flask charged with a stirbar and (2S,3aS,6aS)-2-benzyl 15 tert-butyl hexahydrocyclopenta[b]pyirole-l,2(2H)-dicarboxylate (5.76 g, 16.68 mmol) was added 10% Palladium on carbon (1.77g). Ethanol was poured over the mixture and the reaction mixture was evacuated and flushed with hydrogen gas three times. The suspension was stirred at room température under and atmosphère of hydrogen for 24 hours. Upon completion, the reaction mixture was filtered through celite and concentrated to give (2S,3aS,6aS)-l-(tert-butoxycarbonyl)octahydrocyclopenta[b]pynOle-2-carboxylic acid (4.45g, >99%). MS (ESI) m!z 256.21 [M + H]+.
ντΡ Β Ο-ν «C (2S8 aS,6aS)-benzy 1 octahydrocyclopcnta[b]pyrrolc-2carboxylate hydrochloridc
Intermediate 13
(2S,3S)-2( methoxycarbonylamino}3-metnylpentanolc acid
Pd/C, H2(exccss)
EtOAc
(2S,3aS,6aS)-benzy11-«2S,3S)2-tmethoxy carbonyl a mino)-3m ethyl pentanoyljoctahydrocy do p enta[h]pyrrole-2-carboxylate (2S,3aS,6aS)-1-((2S,3S)-2( methoxycarbonylami no)-3methy I pentanoyl )octahydrocydopenta[t>] pyrrole-2-carboxylic aad (2S,3aS,6aS)-benzyl l-((2S3S)-2-{methoxycarbonyIamino)-3methylpentanoyl)octahydrocyclopenta[bIpyrrole-2-carboxyIate
To a solution of commercially available (2S,3aS,6aS)-benzyl octahydrocyclopenta[b]pyrrole-2-carboxylate hydrochloride (10.0g, 35.489mmo!) in methylene chloride (lOOmL) was added (2S,3S)-2-(methoxycarbonylamino)-320 methylpentanoic acid (10.072g, 53.23mmol), HATU (21.59g, 56.78mmol), and DIPEA (18.59mL, 106.46mmol). The réaction was stirred ovemight, at which time it was concentrated in vacuo, diluted in ethyl acetate and washed with HCl (IN). The aqueous layer was backextracted with ethyl acetate, and the combined organics were dried over sodium sulphate, filtered and concentrated. The resulting oil was diluted in a small amount of chloroform and filtered to remove tetramethyl urea precipitate. The resulting oil was purified by normal phase chromatography (50% ethyl acetate in hexanes) to give (2S,3aS,6aS)-benzyl l-((2S,3S)-2-(methoxycarbonylamino)-3-methylpentanoyl)octahydrocyclopenta[b]pyrrole-2carboxylate (19.53g, >99% yield) which was used without further purification. LCMS-ESI+ calc'd for C23H33N2O5:417.23 ; Found: 417.37.
(2S3aS,6aS)-l-((2S3S)-2-( met h oxyca rbonyla m i no)-3m et hylpen tanoyl)octahyd rocy cio pen ta [ b] pyrroïe-2-ca rbo xy I ic acid
To a 250mL round bottom flask charged with a stirbar and (2S,3aS,6aS)-benzyl 1((2S,3S)-2-(methoxycarbonylamino)-3-methylpentanoyl)octahydrocyc!openta[b]pyrrole’2carboxylate (19.53g crude, assumed 35.49mmol) was added 10% Palladium on carbon (3.55g). Ethanol was poured over the mixture and the reaction mixture was evacuated and flushed with hydrogen gas three times. The suspension was stirred at room température under and atmosphère of hydrogen for 3 days. Upon completion, the reaction mixture was filtered through celite and concentrated to give (2S,3aS,6aS)-l-((2S,3S)-2(methoxycarbonylamino)-3-methylpentanoyl)octahydrocyclopenta[b]pyrrole-2-carboxylic acid (13.65g, >99%). LCMS-ESI+ calc'd for C16H26N2O5: 327.18 ; Found: 327.13.
Intermediate 14
O (2 S,3aS,6a S)-1 -((S}-2-(methoxycartx) ny la mino)-3meUiylbutanoyDoctahydrocyclopenta[b]pynOle-2carboxylic acid (2S3aS,6aS)-l-((S)~2-(methoxycarbonyIamÎno)-3methyIbutanoyl)octahydrocyclopenta[b]pyrrole-2-carboxylie acid (2S,3aS,6aS)-l-((S)-2-(methoxycarbonylamino)-3methylbutanoyl)octahydrocyclopenta[b]pyrrole-2-caiboxylic acid was synthesized in a similar manner as (2S,3aS,6aS)-l-((2S,3S)-2-(methoxycarbonylamino)’3 methylpentanoyl)octahydrocyclopenta[b]pyrTole-2-carboxylic acid substituting (2S,3S)-2(methoxycarbonylamino)-3-methylpentanoic acid with (S)-2-(methoxycarbonylamino)-3methylbutanoic acid. LCMS-ESI+ calc'd for C15H25N2O5: 313.17 ; Found: 313.12.
Intermediate 15
HO
O (2S,5S>1-((S)-2-((2R,6R>2,6-dimettiyttetrahydro2H-pyran-4-yl}-2-(methoxycarbonylamino)acetyl)-5methylpyrrolidine-2-carboxylic acid (2S,5S)-l-((S)-2-((2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2(methoxycarbonyIamino)acetyI)-5-methylpyrrolidine-2-carboxylic acid (2S, 5S>l-((S)-2-((2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-210 (methoxycarbonylamino)acetyl)-5-methylpyrrolidine-2-carboxylic acid was synthesized in a similar manner as Intermediate 4 substituting (S)-2-(methoxycarbonylamino)-3methylbutanoic acid with (S)-2-((2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2(methoxycarbonylamino)acetic acid. 1H NMR (400 MHz, Chloroform-d) δ 5.33 -5.16 (m, 1H), 4.70-4.59 (m, 1Η),4.54(ζ 1 H), 4.34 - 4.19 (m, 2H), 4.12(q, 1H), 3.78-3.70 (m, 1H),
3.67 (s, 3H), 2.37 - 2.17 (m, 3H), 2.15 - 2.07 (m, 1H), 2.04 (s, 1H), 1.84 - 1.73 (m, 1H), 1.82
- 1.43 (m, 3H), 1.32 (d, 3H), 1.26 (d, 4H), 1.11 (d, 3H), 0.96 (q, 1H). LCMS-ESI+ calc'd for
C17H29N2O6: 357.19 ; Found: 357.08.
Example AA
K2CO3
9-bromo-3-(2-bro moacetyf )-10,11 dihydro-5H-di benzo[c,g}chromen6(9H)-one (2S,5S)-1-((2S,3S)-2(methoxycarbonylamino)-3-methyfpentanoyf)5-methy1pyrrolidine-2-carboxy1ic acid
(25,55)-1-(tert-butoxycarbonyi)-5methylpyrro[idine-2-carboxy1ic acid (2S,5S)-2-(9-bromo-8-oxo-8,9,10,11 -tetrahydro-5Hdibenzo[c,g}chromen-3-yl)-2-Oxoethyi 1-((2S,3S)-2(methoxycart>ony1amino)-3-methy1pentanoyl)-5methylpyrrolidine-2-carboxytate
HMDS
Propionic Acid (2S,5S)-1-(ert-butyl2-(3-(2-((2S.5S}-H(2S,3S)-2(methoxycarbonytammo)-3-methylpentanoyl}-5-niethylpynolidine-2carbony1oxy)acety1)-e-oxo-8,9,10,11 -tetrahydro-5Hdibenzo[c.p]chromen-9-yl) 5-methyipyrrolîdrne-1,2-dicart>oxylate
Ο
tert-butyl (2S,5S)-2-{9-(2-{(2S,5S)-1 -{N-(methoxycartx>nyl)-Liso!eucyl]-5-methylpyrTolidtn-2-yi}-1 H-imidazol-5-yl)-1,4,5,11 tetrahydroisochromeno^'.S'ie.TlnaphthoEl.Z-dlirnidazol-Z-ylJ-Smethylpyrrolidine-1 -carboxylate
ΜΠΟ2
1) HCl
2) HATU, DIPEA
tert-butyl (2S,5S)-2-I9-(2-t(2S.5S)-1-EN-(methoxycarbonyl)L-isoleucylJ-5-methylpyrrolidir>-2-yl}-1 H-imidazol-5-yl)-1,11dihydroisochromeno[4',3‘:6.7]na phtho[1,2-d]imidazol-2-yl]-5methylpyrro lîdine-1 -carboxylate
O (S)-2-((2R,6R)-2,6-dimethyltetrahydro-2H-pyran4-y!)-2-(methoxycarbonylamÎno)acetic acid
methyl {(2S,3S)-1-[(2S,5S)-2-(5-P-[(2S,5S)-1-{(2S>-24(2R.6R>-2.6-dimethyltetrahydro2H-pyran-4-yt]-2-[(methoxycart>onyl)aniino jacetyf}-5-methylpyrrohdin-2-yf]-1,11* dihydroisochrarneno[4',3‘:6,7yiaphtho[1,2-d]imidazol-9-y1}-1 H-imidazol-2-y I j-5methy!pyrrohdin-1-yl]-3-methyl-1-oxopentan-2-yÎ}carbamate (2S,5S)-2-(9-Bromo-8-oxo-8,9,10,ll-tetrahydro-5H-dibenzoIc,g]chromen-3-yl)-2 oxoethyl !-((2S,3S)-2-(niethoxycarbonyIamino)-3-methyIpentanoyl)-5methylpyrrolidine-2-carboxylate
To a slurry of 9-bromo-3-(2-bromoacetyl)*l 0,1 l-dihydro-5H-dibenzo[c,g]chromen8(9H)-one (4.00 g, 8.88 mmol) in dichloromethane (50 mL) was added (2S,5S)-l-((2S,3S)-2(methoxycarbonylamino)-3-methylpentanoyl)-5-methylpynolidine-2-carboxylic acid (2.80 g, 9.32 mmol) and K2CO3 (1.84 g, 13.31 mmol). The resulting slurry was stirred at room température for 18 h. The reaction was dîluted with dichloromethane and washed with aqueous HCl (0.5 M) and Brine. The aqueous layers were back extracted with dichloromethane (2x), and the combined organic layers were dried over Na2SO4 and concentrated. The crude product was taken directly into the next reaction.
(2S,5S)-l-Tert-butyl 2-(3-(2-((2S,5S}-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methyIpyrro!idine-2-carbonyloxy)acetyl)-8-oxo-8,9,10,11tetrahydro-5H-dibenzo|c,g]chromen-9-yl) 5-methylpyrroIidine-l,2-dicarboxylate
To a solution of (2S,5S)-2-(9-bromo-8-oxo-8,9,10,l l-tetrahydro-5Hdibenzo[c,g]chromen-3-yl)-2-oxoethyl I-((2S,3S)-2-(methoxycaibonylamino)-3methylpentanoyl)-5-methylpyrrolidine-2-carboxylate (5.95 g, 8.88 mmol) in THF (60 mL) was added (2S,5S)-l-(tert-butoxycarbonyl)-5-methylpyTTolidine-2-carboxyIic acid (3.05 g, 13.3 mmol) and CS2CO3 (2.31 g, 7.09 mmol). The resulting solution was heated to 50 °C for 18 h. The solution was cooled to room température and diluted with EtOAc and washed with aqueous HCl (0.5 M). The aqueous layer was backextracted with EtOAc (2x), and the combined organic layers were dried over NaîSOi and concentrated. The crude oil was purified by column chromatography (S1O2,25-100% EtOAc (5% MeOH)/ Hexanes) to provide (2S,5S)-l-tert-butyl 2-(3-(2-((2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyrrolidine-2-carbonyl oxy)acetyl)-8-oxo-8,9,10,11 -tetrahydro5H-dibenzo[c,g]chromen-9-yl) 5-methylpyrrolidine-l,2-dicarboxylate (3.116,43% over 2 steps) as a orange foam. LCMS-ESI+: calc’d for C44H55N3O12: 817.38 (M+); Found:
817.65 (M+).
Tert-butyl (2S,5S)-2-[9-(2-{(2S,5S)-l-[N-(methoxycarbonyI)-L-îsoleucyl|-5methylpy rroIidin-2-yl}-1 IÏ-imidazol-5-yl)-1,4,5,11 tetrahydroisochromeno|4’3’:6,7]naphthoIl,2-d]imidazol-2-yl]-5-methyIpyrrolidine-lcarboxylate
To a solution of (2S,5S)-l-tert-butyl 2-(3-(2-((2S,5S)-l-((2S,3S)-2(methoxycarbonylamino)-3-methylpentanoyl)-5-methylpyrrolidine-2-carbonyloxy)acetyl)-8oxo-8,9,10,1 l-tetrahydro-5H-dibenzo[c,g]chromen-9-yl) 5-methylpyrrolidine-l ,2dicarboxylate (3.116 g, 3.57 mmol) in toluene (35 mL) was added hexamethdisilazane (6.0 mL, 28.7 mmol), and propionic acid (8.0 mL, 107.1 mmol). The solution was heated to 90 °C for 18h and cooled to room température. The solution was diluted with MeOH and basified with a 1:1 mixture ofNH4OH and water. The slurry was extracted with dichloromethane (3x). The combined organic layers were dried over NaiSO-i and concentrated. The crude oil was used directly in the next step. LCMS-ESI+: calc’d for C44H55N7O6: 777.42 (M+); Found: 778.30 (M+H+).
Tert-butyl (2S,5S)-2-[9-(2-{(2S,5S)-l-[N-(inethoxycarbonyI)-L-isoïeucyl]-5methylpyrrolidin-2-yI}-lH-imÎdazol-5-yl)-l,lldihydroisochromeno[4’,3’:6,7Jnaphtho|l,2-d]iniidazo!-2-ylJ-5-methylpyrrolidine-lcarboxylate
To a solution of tert-butyl (2S,5S)-2-[9-(2-{(2S,5S)-l-[N-(methoxycarbonyl)-Lisoleucyl]-5-methylpyrrolidin-2-yl}-lH-imidazol-5-yl)-l,4,5,lltetrahydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-2-yl]-5-methylpyrrolidine-lcarboxylate (2.77 g, 3.5 mmol) in dichloromethane (25 mL) was added MnO2 (9.00 g, 103 mmol). The resulting slurry was stirred at room température for 20 h. The solution was diluted with dichloromethane, filtered through celite, and concentrated. The crude oil was purified by column chromatography (SiO2, 0-5-10% EtOAc/ MeOH) to provide tert-butyl (2S,5S)-2-[9-(2-{(2S,5S}-l-[N-(methoxycarbonyl)-L-isoleucyl]-5-methylpyrrolidin-2-yl}1 H-imid azol-5-yl)-1,11 -dihydroisochro meno[4', 3 6,7]naphtho[ 1,2-d]i mi dazol-2-yl]-5methylpyrrolidine-1-carboxylate (1.10 g, 40% over 2 steps) as a brown foam. LCMS-ESI+: calc’d for C44H53N7O6: 775.41 (M+); Found: 776.37 (M+H+)
Methyl {(2S3S)-l-[(2S,5S)-2-(5-{2-[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin-2-yl]-l,ndihydroisochromeno[4'r3':6,7]naphtho[l^-d]imidazol-9-yl}-lH-imidazol-2-yI)-5methylpyrrolidin-l-yl]-3-methyl-l-oxopentan-2-yI}carbamate
To a solution of tert-butyl (2S,5S)-2-[9-(2-{(2S,5S)-l-[N-(methoxycarbonyl)-LisoleucyI]-5-methylpyrrolidin-2-yl)-lH-imidazol-5-yl)-l,lldihydroisochromeno[4’,3':6,7]naphtho[l,2-d]imidazol-2-y!]-5-methylpyrrolidine-lcarboxylate (0.30 g, 0.39 mmol) in a mixture of dichloromethane (4 mL) and mcthanol (0.5 mL) was added HCl (4M in dioxanes, 1.45 mL, 5.80 mmol). The solution was heated to 40 °C for 1 h and cooled to room température. The solution was then concentrated in vacuo. The resulting solid was dissolved in DMF (3 mL), followed by the addition of (S)-2((2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid (0.11 g, 0.46 mmol), HATU (0.18 g, 0.48 mmol), and diisopropylethylamine (0.3 mL, 1.72 mmol) The resulting solution was stirred at room température for 3 h. Aqueous HCl (6M, 4 drops) was added and the solution was purified by reverse phase HPLC (Gemini column, 10-53% MeCN/H2O/0.1% TFA). The desired fractions were combined, and the organics were concentrated in vacuo. The resulting aqueous solution was basified with saturated NaHCCh to provide a white precipitate. The solid was filtered and dried to provide methyl {(2S,3S)-1 [(2S,5S)-2-(5-(2-[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2[(methoxycarbonyl)amino]acetyl)-5-methylpyrrolidin-2-yl]-l, 11 dihydroisochromeno[4',3':6,7]naphtho[l,2-d]Îmidazol-9-yl}-lH-imÎdazol-2-yl)-5methylpyrrolidin-l-yl]-3-methyl-I-oxopentan-2-yl}carbamate (0.082 g, 23%) as a white powder. LCMS-ES1+: calc’d for C50H62N8O9: 903.08 (M+); Found: 903.84 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.36 - 8.22 (m, 1H), 7.98 - 7.82 (m, 1H), 7.69 - 7.20 (m, 8H), 5.22 - 5.07 (m, 3H), 5.02 (d, 1H), 4.73-4.64 (m, 1H), 4.49 (s, 3H), 4.26-3.97 (m, 4H), 3.79 (d, 1H), 3.72 (s, 1H), 3.63 - 3.52 (m, 4H), 3.47 - 3.32 (m, 1H), 2.61 -2.43 (m, 1H), 2.32- 1.98 (m, 4H), 1.95- 1.78 (m, 1H), 1.79- 1.65 (m, 1H), 1.54 (s,4H), 1.41 (d, 2H), 1.28 - 1.09 (m, 3H), 1.04 (dd, 6H), 0.90 (d, 1H), 0.88 - 0.59 (m, 10H).
Example AB o—
Methyl {(2S)-l-I(2S,5S)-2-(9-{2-[(2S,4S}-l-{(2S)-2-[(2R,6R)-2,6-dimethyItetrahydro-2Hpyran-4-yl]-2-[(methoxycarbonyl)amÎno]acetyl}-4-(methoxymethyI)pyrrolidin-2-yl]-lHimidazol-5-yl}-l,ll-dihydroisochromeno[4',3’:6,7]naphtho[l,2-d]imidazol-2-yl)-5methylpyrrolidin-l-yl]-3-methyH-oxobutan-2-yl}carbaniate
Following Example AA, substituting (2S,4S)-l-(tert-butoxycarbonyl)-4(methoxymethyl)pyrrolidine-2-carboxylic acid for (2S,5S)-l-((2S,3S)-2(methoxycarbonylamino)-3-methylpentanoyl)-5-methylpyrrolidine-2-carboxylic acid and (2S,5S)-l-((S)-2-(methoxycarbonylamino)-3-methyIbutanoyl)-5-methylpynOlidine-2carboxylic acid for (2S,5S)-l-(tert-butoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid, provided methyl {(2S)-l-[(2S,5S)-2-(9-{2-[(2S,4S)-l-{(2S)-2-[(2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-4(methoxymethyl)pyrrolidin-2-yl]-l H-imidazol-5-yl }-l, 11dihydroisochromeno[4,,3,:6,7]naphtho[l,2-d]imidazo!-2-y!)-5-methylpyrrolidin-l-yl]-3methy1-l-oxobutan-2-yi}carbamate (0.25 g). LCMS-ESI+: calc’d for C50H62N8O9: 918.46 (M+); Found: 919.97 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.39 - 8.11 (m, 1H), 8.09 - 7.40 (m, 5H), 7.28 (s, 2H), 5.27 - 4.91 (m, 5H), 4.49 (s, 3H), 4.34 - 4.14 (m, 2H), 4.14 - 4.00 (m, 3H), 3.80 (s, 1H), 3.59 (s, 2H), 3.57 (s, 3H), 3.54 -3.39 (m, 3H), 3.31 (s, 3H),
2.68 - 2.46 (m, 2H), 2.45 - 2.30 (m, 1H), 2.31 - 1.93 (m, 5H), 1.94- 1.81 (m, 1H), 1.50 (d,
2H), 1.46- 1.25 (m, 4H), 1.10(dd, 3H), 1.06 - 0.90 (m, 9H), 0.87(d, 2H).
Example AC o
Methyl {(lS)-l-[(2R,6R>-2,6-dimethyItetrahydro-2H-pyran-4-ylI-2-|(2S^S)-2-ethyl-5-(9{2-[(2S,5S)-l-{(2S)-2-[(methoxycarbonyl)amÎno]-3-methylbutanoyl}-5-methylpyrrolidinZ-ylI-lH-imÏdazol-S-ylJ-hll-dihydroisochromenoP'^'^tîlnaphthofl^-dlimidazoI-ZyI)pyrrolidin-l-yl]-2-oioethyl)carbamate
Following Example AB, substituting (2S,4S)-l-(tert-butoxycarbonyl)-4(methoxymethyl)pynOlidine-2-carboxylic acid for (2S,5S)-l-((S)-2(methoxycarbonylamino)-3-methylbutanoyl)-5-methylpyrrolidine-2-carboxylic acid and (2S,5S)-l-((S)-2-(methoxycarbonylamtno)-3-methylbutanoyl)-5-methylpyrrolidine-2carboxylic acid for (2S,5S)-l-(tert-butoxycarbonyl)-5-ethy!pynOlidine-2-carboxyIic acid, provided methyl {(1 S)-l-[(2R,6R)-2,6-dim ethyl t et rahydro-2H-pyran-4-y 1)-2-((2 S, 5S)-2ethy 1-5 -(9- {2-[(2S,5S)-l - {(2S}-2-[(methoxycarbonyl)amino]-3 -methylbutanoyl} -5methy 1 pyrrolidin-2-y 1]-1 H-imi dazol-5 -yl} -1,11 -d ihydroisochromeno[4',3': 6,7]naphtho[l,2d]imidazol-2-y!)pyrroIidin-l-yl]-2-oxoethyl) carbamate (0.21 g). LCMS-ESI+: calc’d for C50H62N8O8: 902.47 (M+); Found: 904.14 (M+H+). 1H NMR (400 MHz, Methanol-d4) δ 8.33-8.04 (m, 1H), 8.01-7.68 (m, 1H), 7.68-7.37 (m, 14H), 7.32-7.17 (m, 1 H), 5.224.95 (m, 3H), 4.50 (s, 6H), 4.31-3.92 (m, 5H), 3.80 (s, 1H), 3.58 (d, J = 2.9 Hz, 4H), 3.52 3.35 (m, 1 H), 2.56 - 2.39 (m, 1H), 2.31 - 2.11 (m, 2H), 2.12 - 1.88 (m, 3H), 1.88 - 1.76 (m, 1H), 1.76- 1.48 (m, 1H), 1.40 (d, J = 6.7 Hz, 2H), 1.21 (d, J = 7.0 Hz, 2H), 1.12-1.02 (m, 4H), 0.99 (t, J = 7.3 Hz, 1H), 0.93 (d, J = 7.0 Hz, 1H), 0.89 (d, J = 6.6 Hz, 1H), 0.87 - 0.74 (m, 5H), 0.70-0.45 (m, 1H).
Example AD
tert-butÿl (2S,5S)-2-[9-(2-{(2S,5S}-1 -[N-(methoxycarbonyl)-LvalylJ-5-methy1pyrrofidin-2-yl}-1 H-imidazol-5-yI)-1,11dihydroisochromenoI^B'ze.Tlnaphthofl^-cfJimidazo^-yrç-Sm ethyl pyrroRdine-l -carb
1) HCl
2) HATU, DIPEA
(S)-2-((tertbutoxycarbonyl)amino) -2-(4-methyftetrahydro2H-pyran-4-yf)a cetic acid
1)HCI
methyl [(2S)-1 -((2S.5S)-2-[5-(2-((2S,5S)-1 -[(2S)-2-[(tert- f' butoxycarbon^)amino]A(4-methyltefrahydro-2H-pyran-4-yf)acetyl}-5methyfpyrrolidin-2-yf)-1,11 -dihydroîsochromeno[4,,3,Æ,7jnapWhoÎ12d]imidazol-9-yl)-1 H-imidazol-2-yl]-5-methylpynoridin-1 -yî)-3-methyl-1 oxobutan-2-yl]carbamate
DIPEA DMF
methyl [<2S)-1 -{(2S,5S)-2-[5-(2-((2S,5S)-1-[(2S)-2[(methoxycarbonyl)amino]-2-{4-methyltetrahydro-2H-pyran-4-yl)acetyll5-methyl pyrro tld in-2-yl)-1,11dihydro'rsochromeno[4',3’:6,7]naphtho[1,2-d]imjdazol-9-yf}-1 H-imidazol2-yf]-5-methylpyrroltdin-1 -yl)-3-methyl-1 -oxobutan-2-yf]carbamate
Methyl [(2S)-l-{(2S,5S)-2-[5-(2-{(2S^S)-l-[(2S)-2-[(tert-butoxycarbonyI)aminol-2-(4methyltetrahydro-2H-pyran-4-yl)acetylI-5-niethylpyriOlidin-2-yI}-l,ndihydroisochromeno[4’,3':6'7]naphtho[l,2-d]imidazol-9-y!)-lH~imidazol-2-ylI-5methylpyrroIidin-l-yl}-3-methyl-l-oxobutan-2-yl]carbamate
Methyl [(2S)-l-{(2S,5S)-2-[5-(2-((2S,5S)-l-[(2S)-2-[(tert-butoxycarbonyl)ammo]-2(4-methy ltetrahydro-2H-pyran-4-yl)acety I ]-5 -methylpyrrol i d ίη-2-y1} -1,11 dihydroisochromeno[4’,3':6,7]naphtho[l,2-d]imidazol-9-yl)-lH-imidazol-2-yl]-5methylpynOlidin-l-yl)-3-methyl-l-oxobutan-2-yl]carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2(methoxycarbonylamino)-3-methylbutanoyl)-5-methylpynOlidine-2-carboxyfic acid and sub st i tut i ng ( S)-2-((2R,6R)-2,6-dimethy It etrahydro-2H-pyran-4-yl)-2(methoxycarbonylamino)acetic acid with (S)-2-(tert-butoxycarbonylamino)-2-(4methyltetrahydro-2H-pyran-4-yl)acetic acid. MS (ESI)m/z 917.62 [M + H]+.
Methyl |(2S)-l-{(2S^S)-2-I5-(2-{(2S^S)-l-[(2S)-2-[(methoxycarbonyl)amino]-2-(4methyltetrahydro-2H-pyran-4~yl)acetyl|-5-methylpyrrolidin-2-yl}-l,lldihydroisochromeno[4',3’:6,7]naphtho[l,2-d|imidazol-9-yI)-lH-Îmidazol-2-yl|-Smethylpyrrolidin-l-yl}-3-methy!-l-oxobutan-2-yl|carbamate
To as solution of methyl [(2S)-l-{(2S,5S)-2-[5-(2-{(2S,5S)-l-[(2S)-2-[(tertbutoxycarbonyl)amino]-2-(4-methyltetrahydro-2H-pyran-4-yl)acetyl]-5-methylpyrrolidin-2yl}-1,11 -dihydroisochromeno[4’,3':6,7]naphtho[l ,2-d]imidazol-9-yl)-1 H-imidazol-2-yl]-5methylpyrrolidin-l-yl)-3-methyl-l-oxobutan-2-yl]carbamate (0.594g, 0.648mmol) in a mixture of dichloromethane (6.4 mL) and methanol (1.2 mL) was added HCl (4M in dioxanes, 2.4 mL, 9.72 mmol). The solution was heated to 40 °C for 1 h and cooled to room température. The solution was then concentrated in vactio. The resulting solid was dissolved in DMF (3 mL), followed by the addition of methyl chloroformate (0.050 mL, 0.648 mmol) and diisopropylethylamine (0.14 mL, 0.78 mmol). The reaction mixture was stirred at room température for 30 minutes. Upon completion by LCMS monitoring, the solution was purified by reverse phase HPLC (Gemini column, 10-45% MeCN/lhO/0.1% TFA). The desired fractions were lyophilized to give methyl [(2S)-l-{(2S,5S)-2-[5-(2-{(2S,5S)-l-[(2S)2-[(methoxycarbonyl)amino]-2-(4-methyItetrahydro-2H-pyran-4-yl)acetyl]-5methylpyrrolidin-2-yl}-l,ll-dihydroisochromeno[4',3,:6,7]naphtho[l,2-d]imidazol-9-yl)-lHtmidazol-2-yl]-5-methylpyrro!idin-l-yl)-3-methyl-l-oxobutan-2-yl]carbamate (0.057 g.
100
10%). *H NMR (400 MHz, Methanol-d4) δ 8.63 (s, 1H), 8.19 (d, 1H), 8.09 - 7.75 (m, 4H), 7.75 -7.61 (m, 2H), 7.46-7.24 (m, 1H), 5.39-5.24 (m, 2H), 5.23 -5.12 (m, 1H), 4.84 4.60 (m, 2H), 4.44 - 4.22 (m, 1H), 4.22-4.02 (m, 1 H), 3.85-3.62 (m, 8H), 3,60-3.45 (m,
2H), 2.84-2.44 (m, 2H),2.42-2.23 (m, 3H), 2.12- 1.82 (m, 3H), 1.67(d, 2H), 1.56(d,
4H), 1.37 — 1.19 (m, 4H), 1.17 — 1.03 (m, 4H), 0.98 (d, 3H), 0.88 (d, 3H). MS (ESI) m!z
875.53 [M + H]\
Example AE
Methyl [(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-{(2S,4S)-4-ethyl-2-[510 (2-{(2S,5S}-l-[N-(methoxycarbonyl)-L-valyl|-5-methylpyrroIidm-2-yl)-l,lldihydroisochromeno[4'3’:6,7]naphtho[l,2-d]imidazol-9-yI)-lH-imidazol-2-ylIpyrrolidin-l-yl}-2 oxoethyljcarbamate
Methyl [( 1 S)-1 -[(2R,6R)-2,6-dimethyhetrahydro-2H-pyran-4-yl]-2-{(2S,4S)-4-ethyl2-(5-(2- {(2S,5S)-1- [N-(methoxy carbony l)-L-valyl ] -5 -methyl pyrrol id in-2-y1}-1,1115 dihydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-9-yl)-lH-imidazol-2-yl]pyrrolidin-lyl}-2-oxoethyl]carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3-methylpentanoy!)-5-methylpyrrolidine-2carboxylic acid with (2S,4S)-l-(tert-butoxycarbonyl)-4-ethylpyrrolidine-2-carboxylic acid and substituting (2S,5S)-l-(tert-butoxycarbonyl)-5-methy1pyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)-5-methylpyrrolidine-2carboxylic acid. 1H NMR (400 MHz, Methanol-d4) δ 8.48 - 8.29 (m, 1H), 8.10 - 7.91 (m, 1H), 7.86 - 7.23 (m, 5H), 5.35 - 5.15 (m, 2H), 5.07 (t, 1H), 4.46 - 4.26 (m, 2H), 4.25 - 4.07 (m, 2H), 3.91 (s, 1H), 3.66 (d, 5H), 3.52 - 3.37 (m, 1H), 2.74 - 2.41 (m, 2H), 2.40 - 1.89 (m, 6H), 1.75 - 1.34 (m, 7H), 1.33 - 0.75 (m, 18H). MS (ESI) m!z 903.99 [M + H]+.
101
Example AF
Methyl [(lS)-l-[(2R,6R)-2,6-dimethyItetrahydro-2H-pyran-4-yl]-2-{(2S,4S)-4-ethyl-2-[9-(2{(2S,5S}-l-[N-(niethoiycarbonyl)-L-valyl]-5-methylpyrroIÎdin-2-y]}-lH-imidazol-5-yl)-l,lldihydroisochromenoI4',3’:6/7]naphtho[l,2-d]imidazol-2-yl]pyrrolidin-l-yl}-2oxoethyl]carbamate
Methyl [(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-{(2S,4S)-4-ethyl2-[9-(2- {(2S,5S)-1 -[N-(methoxycarbonyl)-L-valyl]-5-methylpyrro!idin-2-yl}-1 H-imidazol-5yl)-l, 11 -dihydroisochromeno[4’,3':6,7]naphtho[l ,2-d]imidazol-2-yl]pyrrolidin-l-yl}-2oxoethyljcarbamate was synthesized in a similar manner as example AA substituting (2S,5S)-
1- ((2S,3S)-2-(methoxycarbonyIamino)-3-methylpentanoyl)-5-methylpyTTolidine-2-carboxylic acid with (2S,5S)-1 -((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)-5-methylpyrrolidine-
2- carboxylic acid and substiuting (2S,5S)-l-(tert-butoxycarbonyl)-5-methylpyrro!idine-2carboxylic acid with (2S,4S)-l-(tert-butoxycarbonyl)-4-ethylpyrrolidine-2-cafboxylic acid. 1H NMR (400 MHz, Methanol-d4) δ 8.34 (d, 1H), 7.94 (dd, 1H), 7.88 - 7.60 (m, 3H), 7.59 7.28 (m, 2H), 5.25 - 5.11 (m, 3H), 4.54 (s, 1H), 4.39 (t, 1H), 4.27 - 4.12 (m, 2H), 4.12 - 4.02 (m, 1H), 3.62 (d, 4H), 3.48 (s, 3H), 3.13 (s, 3H), 2.68 - 2.45 (m, 1H), 2.38 - 2.19 (m, 2H), 2.19-1.83 (m, 4H), 1.70- 1.53 (m,2H), 1.46 (d, 2H), 1.44-1.28 (m, 3H), 1.28-1.13 (m, 1H), 1.10 (d, 3H), 1.07 - 0.87 (m, 12H), 0.85 - 0.77 (m, 1H). MS (ESI) mlz 903.88 [M + H]+.
Example AG
102
Methyl{(lS)-2-[(2S3S)-2-ethyl-5-(9-{2-I(2S^S)-l-{(2S)-2[(methoxycarbonyl)aniino]-3methylbutanoyl}-5-methylpyrrolidin-2-yl]-lH-imida2ol-5-yI]-l,lldihydroisochromeno[4',3’:6,71naphtho[l,2-d]imidazol-2-yl)pyrroIidin-l-yl]-2-ox(>-l|(3R)-tetrahydro-2H-pyran-3-yl]ethyI}carbaniate
Methyl((lS)-2-[(2S,5S)-2-ethyl-5-(9-{2-[(2S,5S)-l-{(2S)2[(methoxycarbonyl)amino]-3-methylbutanoyl}-5-methylpyrrolidin-2-yl]-lH-imidazol-5yl} -1,11-dihydroisochromeno[4',3':6,7]naphtho[ 1,2-d]imidazol-2-yl)pynOlidin-1-yl]-2-oxo-1 [(3R)-tetrahydro-2H-pyran-3-yl]ethyl)carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S}-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyrro!idine-2-carboxylic acid with (2S,5S)-l-((S)-2(methoxycarbonylamino)-3-methylbutanoyl)-5-methylpyrrolidme-2-carboxylic acid; (2S,5S)-
1- (tert-butoxycarbonyl)-5-methylpyrTolidine-2-carboxylic acid with (2S,5S)-l-(tert-butoxy carbonyl)-5-ethyl pyrrolidine-2-carboxylic acid and (S}*2-((2R,6R)-2,6-dimethyltetrahydro2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid with (S)-2-(methoxycarbonylam!no)-
2- ((R)-tetrahydro-2H-pyran-3-y!)acetic acid. ’H NMR (400 MHz, dmso) δ 8.60 (s, IH), 8.25 - 7.43 (m, 7H), 5.23 (s, 2H), 5.13 (m, IH), 5.01 -4.90 (m, IH), 4.59 (s, IH), 4.33 (m, 2H), 4.12-3.43 (m, 14H),3.37(m, IH), 3.08 (m, 2H), 2.39-1.70 (m, 10H), 1.44 (m, 5H), 1.12 (m, 2H), 0.92 (m, 5H), 0.73 - 0.54 (m, 3H) MS (ESI) m!z 875.95 [M + H]+.
Example AH
MethyI{(lS)-2-[(2S,5S)-2-ethyl-5-(9-{2-[(2S,5S)-l-((2S)-2-I(methoxycarbony!) amino]-3methylbutanoyl}-5-methyIpyrrolidm-2-yl]-lH-imidazol-5-yl}-l,Udihydroisochromeno[4',3':6,7]naphtho[l,2-d|iinÎdazol-2-yl)pyrrolidin-l-yl]-2-oxo-l-[(3S)tetrahy d ro-2H-py ran-3-y I] ethyl} ca rbam ate
Methyl {(1 S)-2-[(2S,5S>2-ethyl-5-(9-(2-[(2S,5S)-l-{(2S)-2-[(methoxycarbonyl) amino]-3-methylbutanoyl} -5-methylpyrrolidin-2-yl]-1 H-imidazoI-5-yl }-l,lldihydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-2-yl)pyrrolidin-l-yl]-2-oxo-l-[(3S)tetrahydro-2H-pyran-3-yl]ethyl}carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbony!amino)-3-methylpentanoyl)-5103 methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxycarbonylamino)-3methylbutanoyl)-5-methy)pynOlidine-2-caiboxylicacid; (2S,5S)-l-(tert-butoxycarbonyl)-5methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-(tert-butoxycarbonyl)-5-ethylpyrrolidine2-carboxylic acid and (S)-2-((2R,6R)-2,6-dimethy]tetrahydro-2H-pyran-4-yl)-2(methoxycarbonylamino)acetic acid with (S)-2-(methoxycarbonylamino)-2-((S)-tetrahydro2H-pyran-3-yl)acetic acid. ’H NMR (400 MHz, dmso) δ 8.64 (s, 1H), 8.24 - 7.46 (m, 8H), 5.27 (s, 2H), 5.13 (s, 1H), 4.99 (s, 1H), 4.62 (s, 1H), 4.12 m, 5H), 3.67 - 3.23 (m, 8H), 3.12 (s, 1H), 2.43 - 2.06 (m, 6H), 2.04 - 1.63 (m, 8H), 1.47 (m, 4H), 1.38 - 1.07 (m, 3H), 0.95 (m, 6H), 0.79 - 0.62 (m, 3H). MS (ESI) mfz 875.86 [M + Hf.
Example AI
Methyl {(lS)-2-[(2S,5S)-2-methyl-5-(9-{2-[(2S,5S)-l-{(2S)-2-[(niethoxycarbonyl) amino]3-methylbutanoyl}-5-methyIpyrrOlidin-2-yl]-lH-imidazoI-5-yl}-l,l 1dihydroisochromeno[4'3’:6,7]naphtho{l,2-d|imidazoI-2-yl)pyrrolidin-l-yI]-2-oxo-l|(3R)-tetrahy d ro-2II-pyran-3-yl] ethyl] carba mate
Methyl {(lS)-2-[(2S,5S)-2-methyl-5-(9-{2-[(2S,5S)-l-{(2S)-2-[(methoxycarbonyI) amino]-3-methylbutanoyl} -5-methylpyrrolidin-2-yl]-1 H-imidazol-5-yl} -1,11 dihydroisochromeno[4,,3,:6,7]naphtho[l,2-d]imidazol-2-yl)pyiTolidin-l-yl]-2-oxo-l-[(3R)tetrahydro-2H-pyran-3-yl]ethyl]carbamate was synthesized in a similar manner as example AA substituting (2S,5 S)-1 -((2S,3 S)-2-(methoxycarbonylamino)-3 -methylpentanoyl)-5methyIpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxy carbonylamino)-3methylbutanoyl)-5-methylpyrrolidine-2-carboxylic acid and (S)-2-((2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid with (S)-2(methoxycarbonylamino)-2-((R)-tetrahydro-2H-pyran-3-yl)acetic acid. ’H NMR (400 MHz, dmso) δ 8.60 (s, 1H), 8.25 - 7.46 (m, 7H), 5.23 (s, 2H), 5.11 (m, 1H), 4.96 (s, 1H), 4.64 (m,
2H), 4.16-3.58 (m, 9H), 3.56-3.31 (m, 6H), 3.08 (m, 3H), 2.19 (m, 5H), 1.86 (m, 3H),
1.43 (m, 7H), 1.24 - 0.92 (m, 3H), 0.83 (m, 3H), 0.68 (m, 3H). MS (ESI) m!z 861.45 [M +
H]+.
104
Example AJ
Methyl {(lS)-2-[(2S,5S)-2-methyl-5-(9-{2-[(2S,5S)-l-{(2S)-2-[(methoxycarbonyl) amino]3-methylbutanoyl}-5~methy!pyrrolidin-2-yl]-lH-imÎdazol-5-yl)-l,lldihydroisochromeno[4',3':6,7]naphtlio[l,2-d]imidazol-2-yl)pyiTolidin-l-yI|-2-oxo-l[(3S}-tetrahydro-2H-pyran-3-yI]ethyl}carbamate
Methyl {(lS>2-[(2S,5S)-2-methyl-5-(9-{2-[(2S,5S)-l-{(2S>2-[(methoxycarbonyl) amino]-3-methylbutanoyl)-5-methylpyrrolidin-2-yl]-lH-imidazol-5-yl}-l,lldihydroîsochromeno[4',3’:6,7]naphtho[l,2-d]imidazol-2-yl)pyrro1idin-l-yl]-2-oxo-l-[(3S)tetrahydro-2H-pyran-3-yl]ethyl}carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3-methylpentanoyl)-5methylpyrro!idine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxy carbonylamino)-3methylbutanoyl)-5-methylpynOlidine-2-carboxyIic acid and (S)-2-((2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid with (S)-2(methoxycarbonylamino)-2-((S)-tetrahydro-2H-pyran-3-yl)acetic acid. ’H NMR (400 MHz, dmso) δ 8.74 - 8.44 (m, 1H), 8.26-7.31 (m, 9H), 5.25 (s, 2H), 5.19 - 5.04 (m, 1H), 5.04 4.87 (m, 1 H), 4.77 - 4.48 (m, 1H), 4.44 - 3.73 (m, 2H), 3.66-2.95 (m,4H), 2.29 (s, 8H), 1.83 (s, 7H), 1.46 (m, 12H), 0.85 (m, 5H), 0.72 (m, 3H). MS (EST) m!z 861.41 [M + H]+.
Example AK
Methyl [(2S)-l-{(2S,5S)-2-[9-(2-{(2S,5S)-l-|(2S}-2-[(tert-butoxycarbonyl)amino]-2-(4methyltetrahydro-2H-pyran^t-yl)acetyl]-5-methylpyrrolidin-2-yl)-l H-imidazol-5-yI)-l ,11dihydroisochromeno(4’3*:6»7Inaphtho[1^2-d]imidazol-2-y]]-5-methylpyrTolïdin-l-yl}-3-methyl- l-oxobutan-2-yl[carbamate
105
Methyl [(2S)-1 - {(2S,5 S)-2-[9-(2- {(2S,5S)-1 -[(2S)-2-[(tert-butoxycarbonyl)amino]-2(4-methyltetrahydro-2H-pyran-4-yl)acetyl]-5-methylpynOlidin-2-yl}-lH-imidazol-5-yl)1,1 l-dihydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-2-yl]-5-methylpyrrolidin-l-yl}-3methyl-l-oxobutan-2-yl]carbamate was synthesîzed in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3-methy!pentanoyl)-5methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-(tert-butoxycarbonyl)-5methylpynOlidine-2-carboxylic acid; (2S,5S)-1 -(tert-butoxycarbonyI)-5-methylpyrrolidine-2carboxylic acid with (2S,5S)-l-((S)-2-(methoxy carbonylamino)-3-methylbutanoyl)-5methylpyrrolidine-2-carboxylic acid and (S)-2-((2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4yl)-2-(methoxycarbonylamino)acetic acid with (S/2-(tert-butoxycarbonylamino)-2-(4methy lt etrahydro-2H-pyran-4-yl)acet ic aci d.
Example AL
methyl [(2S)-1-{(2S.5S)-2-[9-(2-{(2S,5S)-1-[(2S}-2-{(tert-butoxy TU(_ _T cart»nyt)amino]-2-(4-methyttetrahydro-2H-pyran-4-yl)acetyf]-5- uikea, i nr, uri2^i2> κι m ethylpyrrolidtn-2-yl}-1 H-imidazol-5-yî)-1,11 -dihydroisoch romeno (4',3':6,7]naphtho[1,2<f]imidazol-2-y^-5-methyfpyrro!idin-1 -yQ3-methy!-1-oxobutan-2-yf]carbamate
Methyl [(2S)-l-i(2S,5S)-2-[9-(2-{(2Sf5S)-l-[(2S)-2-[(methoxycarbonyl)aminoI-2-(4methyltetrahydro-2H-pyran-4-yl)acetyI]-5-methyIpyrrolidin-2-yl}-lH-imidazol-5-yI)1,1 l-dihydroisochromeno[4'T3':6,7Jnaphtho|l,2-d]imidazoI-2-yl|-5-methylpyrrolidin-lyl}-3-methy I-1 -oxobu ta n-2-y I ] ca rba ma te
Methyl [(2S)-1-((2S,5S)-2-[9-(2-[(2S,5S)-l-[(2S)-2-[(tert-butoxycarbonyl)amino]-2(4-methyltetrahydro-2H-pyran-4-yl)acetyl]-5-methylpyrrolidin-2-yl}-lH-imidazol-5-yl)1,11 -di hydroisochromeno [4',3 ':6,7]naphtho[ 1,2-d]imi dazol-2-yl]-5-methy 1 pyrrolid i n -1 -y 1} -3 106 methyl-l-oxobutan-2-yl]carbamate (316 mg, 0.34 mmol) was dissolved in EtOH (3 mL) and HCl (1 mL) was added. The reaction mixture was stirred for 1 h at 60°C and then concentrated under reduced pressure. The crude residue was dissolved in THF (3 mL) and CH2CI2 (3 mL) treated with DIPEA (0.18 mL, 1.0 mmol) and methyl chloroformate (0.0.03 mL, 0.38 mmol). After 1 h, the mixture was diluted with EtOAc and washed successively with water and brine. The organics were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude residue was purified by reverse phase HPLC (Gemini, 15 to 45% ACN/H2O + 0.1% TFA), to afford methyl [(2S>l-((2S,5S)-2-[9-(2-((2S,5S)-l-[(2S)-2[(methoxycarbonyl)amino]-2-(4-methyltetrahydro-2H-pyran-4-yl)acetyl]-5-methylpyrrolidin2-yl}-lH-imidazol-5-yl)-l,U-dihydroisochromeno [4',3':6,7]naphtho[l,2-d]imidazol-2-yl]-5methylpyrrolidin-l-yl}-3-methyl-l-oxobutan-2-yl]carbamate (100 mg, 33%). ’H NMR (400 MHz, dmso) δ 8.67 (s, 1H), 8.26-7.47 (m, 8H), 5.27 (m, 2H), 5.15 (m, 1H), 5.02-4.90 (m, 1H), 4.70 (s, 1H), 4.44 (s, 1H), 4.29-3.28 (m, 16H), 2.21 (m, 5H), 1.75 (m, 3H), 1.49 (m, 6H), 1.35 - 1.05 (m, 3H), 1.02 - 0.86 (m, 4H), 0.83 (m, 3H), 0.72 (m, 3H). MS (ESI) m!z 875.91 [M + H]+.
Example AM
Methyl {(2SH-[(2S,5S)-2-(5-{2-[(2S,5S)-l-{(2S)-2-[(2R,6S)-2,6-dimethyItetrahydro-2H-pyran-4ylJ-2-J(methoxycarbony!)amino]acetyl}-5-methylpyrrolidin-2-yl]-l,lldÎhydroisochromeno|4*3,:6,7]naphtho|l3-d]imidazol-9-yl}-lH-îmidazol-2-yl>-5methy Ipy rrolid in-1 -yl] -3-methy I-1 -0 xobutan-2-y IJ ca rbam aie
Methyl {(2S)-l-[(2S,5S)-2-(5-{2-[(2S,5S)-l-{(2S)-2-[(2R,6S)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5-methy!pyrrolidin2-yl]-1,11 -dihydroisochromeno[4',3':6,7]naphtho[ 1,2-d]îmidazol-9-yl}-1 H-imidazol-2-yl)-5methylpyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl}carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyrro!idine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxy carbonylamino)-3-methylbutanoyl)-5-methylpyrrolidine-2-cafboxylic acid and (S)-2((2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid with
107 (2S)-2-((2R,6S)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino) acetic acid. *H NMR (400 MHz, dmso) δ 8.64 (s, 1H), 8.29 - 7.49 (m, 7H), 5.26 (s, 2H), 5.15 (m, 1H), 5.07-4.91 (m, 1H),4.61 (m, 2H), 4.17-3.29 (m, 16H), 2.43 - 2.02 (m, 8H), 1.83 (s, 2H), 1.47 (m, 5H), 1.36 - 0.76 (m, 12H), 0.71 (m, 3H). MS (ESI) m!z 889.60 [M + H]+.
Example AN
Methyl {(2S)-l-[(2S,5S)-2-(5-{2-[(2S,5S)*l-{(2R)-2-[(2R,6S}-2,6-diinethyltetrahydro-2H-pyran-4yl]-2-[(methoxycarbonyl)aminoIacetyI}-5-methylpyrrolidin-2-yl]-l,lldihydroîsochromeno[4'3':6,7|naphtho[l^-d]imidazoI-9-yl}-lH-ïinidazoI-2-yl)-5methyIpyrro!ÎdÎn-l-yl|-3-methyl-l-oxobutan-2-yl}carbamate
Methyl {(2S)-l-[(2S,5S)-2-(5-{2-[(2S.5S)-l-{(2R)-2-[(2R,6S)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acety!}-5-methylpyrrolidin2-yl]-l,ll-dihydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-9-yl}-lH-imidazol-2-yl)-5methylpyrro!idin-l-yl]-3-methyl-l-oxobutan-2-yl)carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methyIpentanoyl)-5-methylpyrrolidine’2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxy carbonylamino)-3-methylbutanoyl)-5-methy]pyrrolidine-2-carboxylic acid and (S)-2((2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid with (2R)-2-((2R,6S)’2,6-dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbony!amino) acetic acid. *H NMR (400 MHz, dmso) δ 8.59 (m, 2H), 8.27 - 7.25 (m, 6H), 5.23 (m, 2H), 5.06 4.86 (m, 1 H), 4.76-4.21 (m, 3H), 4.12-2.96 (m, 18H), 2.51-1.69 (m, 12H), 1.65 - 1.33 (m, 6H), 1.25 - 0.55 (m, 8H), 0.07 (m, 2H). MS (ESI) m!z 889.69 [M + Hf.
108
Example ΛΟ
Methyl {(2S>-l-l(2S^S)-2-(9-{2-l(2S3S)-l-{(2S>-2-K2R,6S>2,6-dimethyltetrahydro-2H-pyran-4yI]-2-[(methoxycarbonyl)amino]acetyI}-5-methylpyrrolidin-2-yl]-lII-imidazoI-5-yl}l,ll-dihydroisochromenoI4'3':6,7|naphtho[l^-d|imidazol-2-yI)-5-methylpyrrolidin-l-yII-3methyl-l-oxobutan-2-ylJcarbamate
Methyl {(2S)-l-[(2S,5S>.2-(9-{2-[(2S,5S)-l-{(2S)-2-[(2R,6S)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin2-y I]-1 H-imidazol-5-y!)-1,11 -dihydroisochromeno[4',3':6,7]naphtho[ 1,2-d]imidazoI-2-yl)-5methylpyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl)carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methyIpyrrolidine-2-carboxyIic acid with (2S,5S)-l-(tertbutoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid; (2S,5S)-l-(tert-butoxycarbonyl)-5methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2-{methoxycarbonylamino)-3methylbutanoyl)-5-methylpyrrolidine-2-carboxylic acid and (S)-2-((2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid with (2S)-2((2R,6S)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonyl amino)acetic acid *H NMR (400 MHz, dmso) Ô 8.60 (s, 2H), 8.25 - 7.37 (m, 8H), 5.22 (s, 2H), 5.11 (s, 1H), 4.95 (s, 1H), 4.67 (s, 1H), 4.52 (m, 2H), 3.56 (m, 15H), 2.25 (m, 8H), 1.80 (s, 2H), 1.44 (m, 6H), 1.26 - 0.54 (m, 12H) MS (ESI) m/z 889.56 [M + H] +.
109
Example AP
Methyl {(2S)-l-[(2S,5S)-2-(9-{2-[(2S,5S)-l-{(2R)-2-[(2R,6S}-2,6-dimethyItetrahydro-2Hpyran-4-yl]-2-|(methoxycarbonyl)amino]acetyl}-5-methyIpyrrolidin-2-yl]-lH-imidazol-5-yl}l,ll-dihydroisochromeno[4’3’:6,7|naphtho|l,2-d]imidazol-2-yl)-5-methylpyrrolidin-l-yl[-3methyl-l-oxobutan-2-yl)carbamate
Methyl {(2S)-l-[(2S,5S)-2-(9-{2-[(2S,5S)-l-{(2R>-2-[(2R,6S)-2,6dimethy!tetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin2-yl]-lH-imidazol-5-yl) -1,11 -dihydroisochromeno[4’,3'.6,7]naphtho[l,2-d]imidazo!-2-yl)-5methy!pyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl}carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-(tertbutoxycarbonyl)-5-methylpyiTo!idine-2-carboxylic acid; (2S,5S}-1 -(tert-butoxycarbonyl)-5methyIpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxycarbonylamino)-3methylbutanoyl)-5-methy!pyiTo!idine-2-carboxylic acid and (S)-2-((2R,6R)-2,6dimethyItetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid with (2R)-2((2R,6S)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetÎc acid ’H NMR (400 MHz, dmso) δ 8 60 (m, 1H), 8.24 - 7.41 (m, 8H), 5.23 (m, 2H), 5.10 (m, 2H),
4.65 (s,2H), 3.73 (m, 14H), 2.33 (m, 11 H), 1.84 (s, 3H), 1.54-1.30 (m, 5H), 1.29-0.61 (m, 11H), 0.48 (s, 1H). MS (ESI) m!z 890.05 [M + H]+.
Example AQ
Methyl {(2S)-l-[(2S3S)-2-(S-{2-[(2S3S)-l-((2S)-2-I(2R,6R}-2,6-dimethyltetrahydro-2H-pyran-4yI]-2-[(methoxycarbonyl)ammo]acetyI}-5-methylpyrrolidin-2-yl]-l,Π110 dihydroisochromeno|4',3':6,7]naphtho|I,2-d]imidazol-9-yI}-lH-Îmidazol-2-yl)-5methylpyrroIidin-l-yl]-3-methyl-l-oiobutan-2-yl]carbamate
Methyl {(2S)-l-[(2S,5S)-2-(5-{2-[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl)-5-methylpyrrolidin2-yl]-l,ll-dihydroisochromeno[4',3,:6,7]naphtho[l,2-d]imidazol-9-yl)-lH-imidazol-2-yl)-5methylpyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl}carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methy!pentanoyl)-5-methylpyTroIidine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxy carbonylamino)-3-methylbutanoyl)-5-methylpyrrolidine-2-carboxylic acid. *H NMR (400 MHz, dmso) S 8.63 (s, IH), 8.24- 7.44 (m, 8H), 5.25 (s, 2H), 5.14 (s, IH), 4.99 (s, IH), 4.67 (m, 2H), 3.96 (m, 5H), 3.48 (m, 12H), 2.44 - 1.75 (m, 9H), 1.48 (m, 6H), 1.30-1.10 (m, 3H), 1.01 (m, 3H), 0.85 (m, 4H), 0.75 (m, 3H). MS (ESI) m!z 889.58 [M + H]+.
Example AR
O
Methyl {(2S)-l-[(2S^S>2-(5-{2-[(2S^S}-l-{(2R)-2-I(2R,6R}-2,6-dimethyltetrahydro-2H-pyran-
4-yl]-2-I(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin-2-yl]-l,ndihydroisochromenoi4,3,-6,7]naphthoIl^-d]imÎdazol-9-yl}-lH-imïdazol-2-yI)-5niethyIpyrrolidin-l-yl|-3-methyl-1-oiobutan-2-y]Jcarbamate
Methyl {(2S>l-[(2S,5S)-2-(5-{2-[(2S,5S)-l-{(2R)-2-[(2R,6R)-2,6dimethyItetrahydro-2H-pyran-4-yl]-2-[(methoxycaibonyl)amino]acetyl}-5-methylpyrrolidin2-yI]-l,U-dihydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-9-yI)-lH-imidazol-2-yl)-5methylpyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl)carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methyIpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxy carbonylamino)-3-methylbutanoyl)-5-methyIpyrrolidine-2-carboxylic acid and (S)-2((2R,6R)-2,6-dimethy[tetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid with (R)-2-((2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino) acetic acid. ’H NMR (400 MHz, dmso) δ 8.64 (m, IH), 7.67 (m, 7H), 5.36 - 5.12 (m, 2H), 4.99 (s, 1 H), 4.62 (s, IH), 4.38 (s, lH),4.22(s, IH), 4.16-4.02 (m, IH), 4.00-3.84 (m, 1H),3.7O
111
3.09 (m, 12H), 2.24 (m, 5H), 1.84 (s, 2H), 1.60 (s, 1H), 1.44 (m, 4H), 1.33 - 0.36 (m, 19H). MS (ESI) m!z 889.76 [M + H]*.
Example AS
Ό
Methyl {(2S>l-I(2S^S}-2-(9-{2-[(2S^S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4yl)-2-[(methoxycarbonyl)amino]acetyl}-5-metliylpyrrolidin-2-yI]-lH-imidazol-5-yl)l,ll-dihydroisochromeno[4’,3’:6,7]naphtholl,2-d]imîdîizo!-2-yl)-5-methylpyrrolidin-l-yl]-3methyl-1 -oxobutan-2-yI J carbamate
Methyl {(2S)-l-[(2S,5S)-2-(9-(2-[(2S,5S)-l-((2S)-2-[(2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin2-y 1]-1 H-i mi dazol-5 -y 1} -1,11 -dihydro isochromeno[4',3 6,7] naphtho[ 1.2-d]i mi dazol-2-yl)-5 methylpyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl}carbamate was synthesized in a similar manner as example AA substîtuting (2S,5S)-l-((2S,3S}-2-(methoxycarbonylamino)-3-methyl pentanoyl)-5-methylpynOlidine-2-carboxylic acid with (2S,5S)-l-(tert-butoxycarbonyl)-5methylpyrrolidine-2-carboxylic acid and (2S,5S}-l-(tert-butoxycarbonyl)-5methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxycarbonylamino)-3methylbutanoyl)-5-methylpynOlidine-2-carboxylic acid. ’H NMR (400 MHz, dmso) δ 8.65 (s, 1H), 8.23 - 7.44 (m, 8H), 5.26 (s, 2H), 5.15 (s, 1H), 4.99 (s, 1H), 4.66 (m, 2H), 4.24 3.82 (m, 4H), 3.75-3.20(m, 12H), 2.42 - 1.72(m, 10H), 1.47(m, 5H), 1.30-0.96 (m, 6H), 0.95 - 0.62 (m, 8H). MS (ESI) m!z 889.88 [M + H] *.
Example AT
O
Methyl {(2S)-l-I(2Si5S)-2-(9-{2-[(2S,5S)-l-{(2R)-2-|(2R,6R)-2,6-dimethyItetrahydro-2Hpyran-4-yl|-2-[(methoiycarbonyl)amino|acetyl)-5-methy!pyrrolÎdin-2-yl|-lH-îmidazoI-5-yl}
112 l,ll-dihydrotsocliron)eno[4*3':6,7]naphtho[l^î-d|iinïdazol-2-yl}*5-inethylpyrTOIidin-l-y!|-3methyl-l-oxobutan-2-yl|carbamate
Methyl {(2S)-l-[(2S,5S)-2-(9-{2-[(2S,5S)-l-{(2R)-2-[(2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin2-yl]-1 H-ïmidazol-5-y 1} -1,11 -dihydroisochromeno[4',3':6,7]naphtho[ 1,2-d]imidazol-2-yI)-5methylpyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl)carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-(tertbutoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid; (2S,5S)-1 -(tert-butoxycarbonyl)-5methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxycarbonylamino)-3methylbutanoyl)-5-methylpynOlidine-2-carboxy!ic acid and (S)-2-((2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycarbonylamino)acetic acid with (R)-2((2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl)-2-(methoxycafbonyl amino)acetic acid *H NMR (400 MHz, dmso) δ 8.65 (s, 1H), 7.74 (m, 8H), 5.71 - 5.53 (m, 1H), 5.28 (s, 2H), 5.15 (s, 1H), 5.05 (s, 1H), 4.70 (s, lH),4.13(m, 5H), 3.82-3.15 (m, 1 OH), 2.70-2.57 (m, 1H), 2.43- 1.71 (τη, 9H), 1.67- 1.29 (m, 6H), 1.28-0.54 (m, 14H). MS (ESI) m/z 889.53 [M + H]+.
Example AU
Methyl {(lS)-l-[(2R,6R)-2,6-dimethyItetrahydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(5-{2[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dÎmethyltetrahydiO-2H-pyran-4-yl]-2[(methoxycarbonyl)amino]acety!}-5-methyIpyrrolidin-2-yl]-l,lldihydroisochromeno[4’3’:6»7]naplitho[14-dIiniidazoI-9-yI}-lH-imidazol-2-yl)-5methylpy rrolid in-l-yl]-2-oxoethyl} ca rba mate
Methyl {(lS)-l-[(2R,6R)-2,6-dimethy]tetrahydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(5-{2[(2S,5S)-l-((2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2[(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin-2-yl]-l,lldihydroisochromeno[4',3':6,7]naphtho[I,2-d]imidazol-9-y]}-lH-imidazol-2-yl)-5113 methylpynolidin-l-yl]-2-oxoethyl)carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyrrolidine*2-carboxylic acid with (2S,5S)-l-(tertbutoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid. *H NMR (400 MHz, dmso) δ 12.94 5 (s, 1H), 12.36 (s, 1H), 11.77 (s, 1H), 8.42 (m, 1H), 8.13 - 7.16 (m, 7H), 5.11 (s, 3H), 4.96 (s,
1H), 4.64 (s, 2H), 3.97 (m, 4H), 3.67-3.11 (m, 13H), 2.39-1.66 (m, 10H), 1.62- 1.30 (m, 7H), 1.30 - 0.92 (m, 9H), 0.81 (m, 6H). MS (ESI) m!z 960.04 [M + H] +.
Example AU-2
9-bromo-3-(2-bromoacetyl)-10,11 dihydro-5H-dibenzo[c,g]chromen8(9H)-one
CS2CO3 (2S,55)-1 -ftert-butoxycarbonyl)-5methylpyrrolidine-2-carboxylic acid
(2S,5S)-2-(2-(9-{((2S,5S)-1-(tert-butoxycarbonyl)-5-methylpyrrolidine2-carbony1)oxy)-8-oxo-8,9,10,11-tetrahydro-5/7-dibenzo[c,g]chromen3-y1)-2-oxoethyl) 1-tert-butyl 5-methylpyrrolicfine-12-di carboxylate
NH4OAC isopropanol
(2 S,5 S)-tert-buty! 2-(5-(2-((25,5 S}-1 -(t erf-butoxyca rbonyl)-5methylpyrrolidin-2-yl)-1,4,5,11-tetrahydroisochromeno[4*,3‘:6,7lnaphtho[12d]imidazol-9-yl)-1 H-imidazol-2-y1)-5-methyÎpynOÎÎdine-1 -carboxylate
MnO2
114
(2S,5S)-tert-buty12-(5-(2-((2S,5S}-1-(tert-butoxycarbonyI)-5methylpyTTolidin-2-yl)-1,11-dihydroisochromeno[4’,3‘:6,7]naphtho[1,2d]imidazol-9-yi)-1 H-imidazol-2-y1)-5-methylpyrroîîdine-1 -carboxylate
HCl
2-((2S,5S)-5-methylpyrTofidin-2-yi)-9-(2-{(2S,5S>-5methy1pyrTolidin-2-yi)-1 H-imidazol-5-yl)-1,11dihydroisochrorneno[4’,3':6,7]naphtho[1 ,2-d]imidazole
( S)-2-((2R,6R}-2,6-dimethy Itetra hydro-2H-pyra n4-yl)-2-((methoxycarbonyl)amino)aceticacid
Methyl {(1S)-1-{(2R,6R}-2,6-dimethyltetrahydro-2H-pyran-4-yl}-2-((2S,5S)-2-(5-{2-[(2S,5S)-1{(2S)-24(2R,6R)-216-dimethyttetrahydro-2H-pyran-4-yl}-2-{(methoxycarbonyl)amino]acetyÎ}-5methylpyrrolidin-2-yQ-l ,11 -dihydrotsochromenoH’.TÆ.Tlnaphthotl ,2-d]imidazo!-9-yf}-1 Hi midazol-2-yl)-5-methylpynoli din-1 -y1]-2-ox oethyQcarba mate (2S,5S)-2-(2-(9-(((2S,5S)-l-(tert-butoxycarbonyl)-5-metliylpyrrolidine-2carbonyl)oxy)-8-oxo-8,9,10,n-tetrahydro-5H-dibenzo[c,gIchromen-3-yl)-2-oxoethyl) 15 tert-butyl 5-methylpyrrolidine-l^-dïcarboxylate
To a solution of (2S,5S)-l-(tert-butoxycarbonyI)-5-methylpyrrolidine-2-carboxy!ic acid (25.44 g, 111 mmol) in THF (400 mL) was added césium carbonate (21.7 g, 66.6 mmol). The solution was stirred at room température for 15 min, followed by the addition of 9bromo-3-(2-bromoacetyl)-10,l l-dihydro-5H-dîbenzo[c,g]chromen-8(9H)-one (19.9 g, 44.4 10 mmol). The slurry was stirred at room température for 15 minutes and then heated to 40 °C for 16 hours. The reaction was cooled to room température and diluted with EtOAc. The solution was washed with 0.3 M aqueous HCl. The aqueous layer was backextracted with EtOAc, and the combined organic layers were dried over Na2SOi and concentrated. The
115 crade oïl was then fîltered through a plug of silica gel (260g) with 40% EtOAc/Hexanes. The filtrâtes were combined and concentrated to provide (2S,5S)-2-(2-(9-(((2S,5S)-l-(tertbutoxycarbonyl)-5-methylpyrT01idine-2-carbonyl)oxy)-8-oxo-8,9,10,11 -tetrahydro-5Hdibenzo[c,g]chromen-3-yl)-2-oxoethyl) 1-tert-butyl 5-methylpyrrolidine-l,2-dicarboxylate (33g, 100%). MS (ESI) m!z 647.57 [M - Boc] * (2S^S)-tert-butyl 2-(5-(2-((2S,5S)-l-(tert-butoxycarbonyl)-5-inethylpyrrolidin-2yl)-l,4,5,ll-tetrahydroisochronieno[4’T3,:6,7]naphtho[l,2-d]ÎmidazoI-9-yl)-lH-imidazol-
2-yl)-5-methyIpyrrolidine-l-carboiylate
To a solution of (2S,5S)-2-(2-(9-(((2S,5S)-l-(tert-butoxycarbonyl)-5methylpyrrolidine-2-carbonyl)oxy)-8-oxo-8,9,10,11 -tetrahydro-5H-dibenzo[c,g]chromen-3yl)-2-oxoethyl) 1-tert-butyl 5-methylpyrroIidine-l,2-dicarboxylate (49.3 g, 66 mmol) in toluene (600 mL) and isopropanol (66 mL) was added ammonium acetate (102 g, 1323 mmol). The reaction was heated to 90 °C for 18 hours and cooled to room température. The reaction was quenched by the addition of water (200 mL) and stined at room température for 10 min. The organic layer was isolated and washed with water (200 mL). The combined aqueous layers were backextracted with a mixture of toluene/isopropanol/methanol (10:1:1, 60 mL). The combined organic layers were transferred into a roundbottom flask and celite (29 g), methanol (150 mL), and a 2:1 mixture ofbrine and 6M NaOH were added. After vigorous stirring for 40 min, the slurry was fîltered and rinsed with a 9:1 mixture of toluene/isopropanol. The organic layer was isolated, diluted with EtOAc, and washed with water. The organic layer was then concentrated, dissolved in a mixture of CHîClî/hexanes, and concentrated to dryness. This provided (2S,5S)-tert-butyl 2-(5-(2-((2S,5S)-l-(tertbutoxycarbonyl)-5-methyIpyrro!idin-2-yl)-l,4,5,l 1t etrahydroi sochrom eno[4',3': 6,7] naphthof 1,2-d] imi dazol-9-yl)-1 H-i midazol-2-y l)-5methylpyrrolidine-l-carboxylate (32.3 g, 69%). MS (ESI) m!z 707.30 [M + H]+ (2S,5S)-tert-butyl 2-(5-(2-((2S^S)-l-(tert-butoxycarbonyl)-5-methylpyrrolidin-2yl)-l,ll-dihydroisochromeno[4'T3,:6,7]naphthoJl,2-d|imidazol-9-yl)-lH-imidazol-2-yI)-
5-methylpyrrolidine-l-carboxylate
To a solution of (2S,5S)-tert-butyl 2-(5-(2-((2S,5S)-l-(tert-butoxycarbonyl)-5methylpyrrolidin-2-yl)-l,4,5,ll-tetrahydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-9yl)-lH-imidazol-2-yl)-5-methylpyrroIidine-l-carboxylate (32.3 g, 45.7 mmol) in CH2CI2 (420 mL) was added manganèse dioxîde (120 g, 1380 mmol). The slurry was stined at room
116 température for 46 hour. The reaction was then diluted with CH2CI2 and celite was added. After stirring at room température for 20 min, the slurry was filtered and the celite was washed with CH2CI2. The filtrate was concentrated to provide (2S,5S)-tert-butyl 2-(5-(2((2S,5S)-1 -(tert-butoxycarbonyl)-5-methylpyrrolidin-2-yI)-l, 11 dihydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-9-yl)-lH-imidazol-2-yl)-5methylpyrrolidine-l-carboxylate (25.1 g, 78%). MS (ESI) m!z 705.50 [M + H]+
2-((2S,5S)-5-methylpyrrolidin-2-yl)-9-(2-((2S,5S)-5-methylpyrrolidin-2-yI)-lHimidazoI-5-yl)-l,ll-dihydrOisochrOmeno[4’r3’:6,7]naphtho[l,2-d]imidazoIe
To a solution of (2S,5S)-tert-butyl 2-(5-(2-((2 S,5 S)-l-(tert-butoxycarbonyl)-5methylpyrrolidin-2-yl)-l, 11 -dihydroisochromeno[4',3':6,7]naphtho[l ,2-d]imidazol-9-yl)-l Himidazol-2-yl)-5-methylpyrrolidine-l-caTboxylate (25.1 g, 35.6 mmol) in CH2CI2 (350 mL) was slowly added HCl (6M in dioxane, 180 mL, 720 mmoL) over 5 minutes. The solution was stirred at room température for 30 minutes, and then warmed to 40 °C for 1.5 hours. The resulting slurry was cooled to room température and filtered. The precipitate was then washed with CH2CI2 and dried to provide 2-((2S,5S)-5-methylpyiToiidin-2-yi)-9-(2-((2S,5S)5-methylpyrrolidin-2-yl)-lH-imidazol-5-yl)-l,ll-dihydroisochromeno[4',3*:6,7]naphtho[l,2djimidazole as the tetra-HCl sait (21.2 g, 92%). MS (ESI) m!z 505.18 [M + H]+
Methyl {(lS)-l-|(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(5{2-[(2S^S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-y!]-2[(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin-2-yI]-l,l 1dihydroi30chromeno[4*3':6,7|naphtho[I,2-d]imîdazol-9-yl}-lH-imidazoI-2-yI)-5methylpyrroïidin-l-yl]-2-oxoethyl}carbamate
To a solution of l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDCI.HC1, 11.7 g, 61 mmol) in DMF (180 mL) was added 1-hydroxybenzotriazole (8.2 g, 61 mmol). The slurry was stirred until homogeneous and cooled to 2 °C. (S)-2-((2R,6R)2,6-dimethyltetrahydro-2H-pyran^4-yl)-2-((methoxycarbonyl)amino)acetic acid (15.0 g, 61.2 mmol) was added and stirred at 1 °C for 1 hour. Then 2-((2S,5S)-5-methylpyrrolidin-2-yl)-9(2-((2S,5S)-5-methylpyrrolid!n-2-yl)-lH-imidazol-5-yl)-l,l 1dihydroisochromeno[4*,3';6,7]naphtho[l,2-d]imidazole tetra-HCl sait (15.6 g, 27.1 mmol) and n-methylmorpholine (17.8 mL, 162.6 mmol) were added. The resulting mixture was stirred at room température for 22 hours. The reaction was then diluted with EtOAc and wash with NH4CI (2x 500 mL), aqueous bicarbonate (2x 300 mL), and brine (300 mL). The aqueous
117 layers were backextracted with EtOAc. The combined organic layers were dried over NaîSOi and concentrated. The crade material was purified through a plug of silica gel with 3 to 8% MeOH/CHzCh to provîde methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran4-yl]-2-[(2S,5S)-2-(5-{2-[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4yl]-2-[(methoxycarbonyl)amino]acetyl} -5-methyl pyrrolidin-2-yl]-1,11dihydroisochromeno[4',3’:6,7]naphtho[l,2-d]imidazol-9-yl}-lH-imidazol-2-yl)-5methylpyrrolidin-l-yl]-2-oxoethyl)carbamate (20.5 g, 79%). MS (ESI) mtz 959,87 [M + H] +
Example AV
methyl {(2S)-l-((2S,5S)-2-(5-{2-[(2S,4S}-l-{(2S)-2-|(2R,6R)-2,6-dimethyltetrahydro-2Hpyran-4-yl]-2-[(methoxycarbony!)amino]acetyI}-4-inethylpyrrolidin-2-yl]-l,l 1dihydrOisochromeno[4'r3':6,7Inaphtho[l,2-dlimidazoI-9-yI}-lII-imidazoI-2-yl)-5methy!pyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yI}carbamate
Methyl {(2S)-l-[(2S,5S)-2-(5-{2-[(2S,4S)-l-{(2S)-2-[(2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-4-methylpyTrolidin2-yl]-l, 11-dihydroisochromeno[4',3';6,7]naphtho[ 1,2-d]imîdazol-9-yI }-l H-imidazol-2-yl)-5methylpyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl}caibamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyTrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2(methoxycarbonylamino)-3-methylbutanoyl)-5-methylpyiTolidine-2-carboxylic acid, and substiuting (2S,5S)-l-(tert-butoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid with (2S,4S)-l-(tert-butoxycarbonyl)-4-methyIpynOlidine-2-carboxylic acid. ’H NMR (400 MHz, Methanol-^) δ 8.63 (d, 1H), 8.19 (d, 1H), 8.11 - 7.76 (m, 4H), 7.72 - 7.57 (m, 1H), 5.44 5.26 (m, 2H), 5.23 - 5.11 (m, 1 H), 5.00-4.71 (m, 5H), 4.47 (t, 1 H), 4.16 (dt, 3H), 3.81 3.62 (m, 5H), 3.53 (t, 1H), 2.83 - 2.67 (m, 1H), 2.53 (dd, 2H), 2.33 (dd, 2H), 2.04 (dd, 4H), 1.54 (dd, 2H), 1.52- 1.38 (m, 3H), 1.28 (d, 3H), 1.20 (s, 1H), 1.15 (s, 2H), 1.11-0.95 (m, 6H), 0.87 (t, 2H). LCMS-ESI+ calc’d for C49H61N8O8 : 890.05 ; Found: 889.23.
118
methyl {(2S)-l-[(2S,5S>-2-(5-{2-[(2S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2tIpyran-4~yl]-2-[(methoxycarbonyl)amino]acetyI}pyrroïidin-2-yl]-l,lldihydroisochr(>meno[4*,3':6,7]naphtho[l,2-d]imidazol-9-yl}-lH-imidazol-2-yl)-5m ethy Ipy rrol id in-1 -y I]-3-met hy I-1 -oxobu ta n-2-y 1} ca rbamate
Methyl {(2S)-l-[(2S,5S)-2-(5-{2-[(2S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro2H-pyran-4-y l]-2- [(methoxy carbony l)ami nojacetyl} pyrrol id in-2-y 1 ]-1,11d ihydroi sochromeno [4',3’: 6,7]naphtho[ 1,2-d]imi dazol-9-y I} -1 H-îmî dazol-2-yl)-5methylpyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl}carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S}-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpynOlidine-2-carboxylic acid with (2S,5S)-l-((S)-2(methoxycarbonylamino)-3 -methy lbutanoyl)-5 -methyl pyrrol idi ne-2-carboxy 1 îc acid, and substiuting (2S,5S)-l-(tert-butoxycaTbonyl)-5-methylpynO!idine-2-carboxylic acid with (S)l-(tert-butoxycarbonyl)pyTTolidine-2-carboxylic acid. 1H NMR (400 MHz, Methanol-d4) δ
8.65 (s, 1H), 8.21 (d, 1H), 8.08 - 7.96 (m, 1H), 7.92 (s, 1H), 7.85 (d, 1H), 7.75 - 7.59 (m, 2H), 5.45-5.38 (m, 1H),5.33 (s, 1H), 5.23 - 5.11 (m, 1H), 4.35-4.06 (m, 4H), 4.01-3.92 (m, 1H), 3.82 - 3.44 (m, 7H), 2.75 - 2.48 (m, 3H), 2.46 - 2.09 (m, 6H), 2.07 - 1.91 (m, 2H), 1.56 (d, 3H), 1.49- 1.36 (m, 2H), 1.32-1.21 (m,2H), 1.15 (d,3H), 1.10-0.93 (m, 6H), 0.88 (d, 2H).. LCMS-ESI+ calc’d for C48H59N8O8 : 875.45 ; Found: 875.29.
119
Exemple ΑΧ
Ο methyl {(2S>l-I(2S,5S)-2-(5-{2-[(2S3aS,6aS)-l-{(2S)-2-[(2R,6R>-2,6d im ethy Itet rahy d ro-211-py ra n-4-yI]-2[(methoxycarbonyl)aminoJacetyl}octahydrOcyc!openta[bJpyrroI-2-yl]-l,lldihydroÎsochromeno[4’3':6,7]naphtho[l,2-d|imidazoI-9-yl}-lH-imidazol-2-yI)-5methylpyrrolidin-l-ylJ-3-methyl-l-oxobutan-2-yl}carbamate
Methyl {(2S>l-[(2S,5S)-2-(5-{2-[(2S,3aS,6aS)-l-{(2S)-2-[(2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2[(methoxycafbonyl)amino]acetyl}octahydrocyc!openta[b]pyrro!-2-yl]-l,l 1dihydroisochromeno[4',3’:6,7]naphtho[l,2-d]imidazol-9-yl}-lH-imidazol-2-yl)-5methylpyrrolidin-l-yl]-3-methyl-l-oxobutan-2-yl)cafbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamÎno)-3methylpentanoyl)-5-methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2(methoxycarbonylamino)-3-methylbutanoyl}-5-methylpyiTolidine-2-carboxylic acid, and substiuting (2S,5S)-l-(tert-butoxycarbonyl)-5-methylpynOlidine-2-carboxylic acid with (2S,3aS,6aS)-l-(tert-butoxycarbonyl)octahydrocyclopenta[b]pyrrole-2-carboxylic acid. 1H NMR (400 MHz, Methanol-d4) δ 8.66 (s, 1H), 8.21 (d, 1 H), 8.12 - 7.97 (m, 2H), 7.94 (s, 1H), 7.91 - 7.78 (m, 1H), 7.74 - 7.62 (m, 2H), 5.81 - 5.71 (m, 1H), 5.39 - 5.26 (m, 3H), 5.23 - 5.11 (m, 1H), 4.84 -4.76 (m, 2H), 4.25 (d, 1H), 4.21-4.06 (m, 2H), 3.81 (s, 1H),3.74 (s, 1H), 3.67 (d, 4H), 3.63 - 3.52 (m, 1H), 3.16-3.04 (m, 1H), 2.76 - 2.64 (m, 1H), 2.61 - 2.48 (m, 1H), 2.44 - 2.29 (m, 3H), 2.23 (q, 2H), 2.18 - 2.08 (m, 2H), 2.07 - 1.85 (m, 4H), 1.84 - 1.66 (m, 2H), 1.57 (d, 3H), 1.49 - 1.36 (τη, 2H), 1.29 (t, 1H), 1.27 - 1.18 (m, 1H), 1.15 (d, 3H), 1.12-1.01 (m, 4H), 0.98 (d, 3H), 0.88 (d, 2H). LCMS-ESI+ calc’d for C51H63N8O8 : 915.48; Found: 915.29.
120
Example A Y
methyl {(2S>l-[(2S3aS,6aS)-2-(5-{2-I(2S3S)-l-((2S)-2-[(2R,6R>-2,6dimethyltetrahydro-2II-pyran-4-yl]-2-[(methoxycarbonyl)amino]acctyl}-5methylpyrrolidin-2-yl]-l,ll-dihydroisochromeno[4'3':6,7]naphtho[l,2-d]imidazoî-9yI}-lH-imidazoI-2-yl)hexahydrocyclopenta[b]pyrroI-l(2II)-yl]-3-methyl-l-oiobutan-2yljcarbamate
Methyl {(2S)-l-[(2S,3aS,6aS)-2-(5-{2-[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5-methylpynOlidin2-yl]-l,ll-dihydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-9-yl}-lH-imidazol-2yl)hexahydrocyclopenta[b]pyrro!-l (2H)-yl]-3-methyl-l -oxobutan-2-yl} carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2(methoxy carbony Iamino)-3 -methyl pentanoy I)-5-methyl pyrroli di nc-2-carboxy 1 ic acid with (2S,3aS,6aS)-l -((S)-2-(methoxycarbony lam ino)-3methy!butanoyl)octahydrocyclopenta[b]pyiTole-2-carboxylic acid, 1H NMR (400 MHz, Methanol-d4) δ 8.66 (s, IH), 8.29 - 8.18 (m, 1H), 8.10 - 8.03 (m, 1H), 7.94 (d, 1H), 7.84 (d, 1H), 7.73 - 7.64 (m, 1H), 5.32 (d, 2H), 5.24 - 5.13 (m, 1H), 4.18 (dd, 2H), 3.86 - 3.72 (m, 2H), 3.67 (d, 3H), 3.52 (d, 1H), 3.11 - 2.97 (m, 1H), 2.72 - 2.57 (m, 2H), 2.54 - 2.21 (m, 3H), 2.20 - 1.80 (m, 7H), 1.79- 1.70 (m, 1H), 1.65 (d, 2H), 1.61-1.52 (m, 1H), 1.50- 1.36 (m, 1H), 1.36- 1.23 (m,2H), 1.14 (d,3H), 1.10- 1.02 (m, 1H), 1.02-0.88 (m, 6H).. LCMSESI+ calc'd for C51H63N8O8 :915.48 ; Found: 915.379.
121
Examples AZ, BA, BB, BC
methyl {(1S)-1-{(2R.6R)-2l6-dmethyltetrahydro-2H-pyrarM-yf]-2-[(2S,3S,5S)5-(5-{2-[(2S,4S>5S)-1-{(2S)-2-{(2R,6R)-2,6-dmethyltetrahydro-2H-pyran-4-yil2-4(methoxycarbonyl)amino]acetyl}-4,5-dimethylpyrrolidin-2-yl}-1,11’ d hydroisochromeno[4'l3'.6,7] naphtho[1,2-d]i mtdazol-9-y1}-1 H-imidazol-2-yl)-
2,3-dimethylpyrrolid n-1-ylJ-2-oxoethy1}ca rba mate
methyl {(1S)-1-[(2R,6R)-2,6-dimethyttetrahydro-2H-pyran-4-yî}-2-[(2Rl3R,5R)5-(9-{2-((2S,4Sl5S)-1-{(2S)-2-[(2R.6R)-2,6-dimethyltetrahydro-2H-pyran-4-ylj2-{(methoxy carbonyl)amino]acetyl}-4,5-d methyl pyrrolidn-2-yl]-1 H-ι midazol-5ylj-1,11-dit^drasochromeno[4‘,3'.6,7]naphtho[1,2-d]imidazol-2-yl)-2,3dimethylpyrrolidîn-1-yl]-2-oxoethyI}carbamate
methyl {(1S)-1-i(2R, 6R)-2,6-dimethy1tetrahydro-2H-pyran-4-y1}-2-{(2R,3R,5R)545-{2-{(2Sl4S,5S)-14(2S)-2-{(2R,6R}-2,6-dirB^yitetrahydro-2H-pyran-4-ylJ2-{(methoxycarbonyl)amino]acetyf}-4,5-d methyl pyrrolicün-2-y1]-1,11 di hydroisochromeno[4'13‘. 6,7]naphtho[1,2-d]i midazol-9-yt}-1 H-imidazol-2-yl)-
2,3-d methylpy rrolid n-1 -yl]-2-oxoethyl}ca rbamate
methyl {(1S)-1-[(2R.6R)-2,6-cimethyltetrahydro-2H-pyran^-y1]-2-{(2R.3R,5R)5-{5-{2-{(2R,4Rl5R)-1-{(2S)-2-{(2Rl6R)-2l6-dimethyttetrahydro-2H-pyran-4-yl]2-((methoxycartxjnyl)amino]acety!}-4,5-<fimethylpyrrolidn-2-yl>1,11’ d hydroisoch romen 0(4^:6,7] na phtho[1,2-d]i midazol-9-yl}-1 H-imidazol-2-yl )-
2,3-dtmethylpyrrolidin-1-yl}-2-oxoethyf}carba mate
122 methyl {(lS)-l-[(2R,6R)-2,6-dimethyItetrahydro-2H-pyran-4-y!]-2-[(2S3S,5S)-5-(5-{2[(2S,4S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-ylJ-2[(methoxycarbonyI)amino]acetyI}-4,5-dimethyIpyrroIidin-2-yI]-l,lldihydroisochromeno[4'3’:6,7]naphtho[l,2-d]imidazol-9-yl}-lH-imidazol-2-yl)-23dimethylpyrroIidin-l-yl]-2-oxoethyl}carbamate.
methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(2R3R3R)-5-(9-{2[(2S,4S,5S>l-{(2S>2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2[(methoxycarbonyl)amino]acetyI}-4,5-dimethylpyrrolidin-2-yl]-lH-iinidazol-5-yl}-l,l 1dihydroisochromeno[4*3':6>7]naphtho[l,2-d)imidazol-2-yl)-23-dimethylpyrrolidin-lyl]-2-oxoethy!} ca rbamate methyl {(lS}-l-[(2R,6R)-2,6-dimcthyItetrahydro-2H-pyran-4-yl]-2-[(2R3R,5R)-5-(5-{2[(2S,4S3S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyItetrahydro-2H-pyran-4-yl]-2[(methoxycarbonyI)amino]acetyI}-4,5-dimethylpyrrolidin-2-yl]-l,l Idihydroisochromeno[4’3’:6,7]naphtho[l,2-d|imidazoI-9-yl}-in-ÎmidazoI-2-yl)-23dimethylpyrro!idin-l-yl]-2-oxoethyI}ca rbamate methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yII-2-[(2R3R,5R)-5-(5-{2[(2R,4R,5R)-l-{(2S)-2-[(2R,6R)-2,6-dimethyItetrahydro-2H-pyran-4-yl]-2[(methoxycarbonyl)amino]acetyl}-4,5-dimethylpyrrolidin-2-yl]-l,lldihydroisochromeno|4,3':6,7]naphtho[l,2-d]imîdazoI-9-yl}-lH-imidazol-2-yl)-23' dimethylpyrrolidin-l-yl]-2-oxoethyl}ca rbamate
Following Example AA, substituting rel-(2S,4S,5S)-l-(tert-butoxycarbonyl)-4,5dimethylpyrrolidine-2-cafboxylic acid for (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyrrolidine-2-carboxylic acid and (2S,5S)-l-(tertbutoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid, and using two équivalents of (S)-2((2R,6R)-2,6-di methy ltetrahydro-2H-pyran-4-y l)-2-(methoxycarbony 1 ami no)acet ic acid, provided a mixture of four diastereomers. The diastereomers were separated by reverse phase HPLC (Gemini column, 10-45% MeCN/H20/0.1% TFA).
Example AZ. Methyl {(!S)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yI]-2[(2S,3S,5S)-5-(5-{2-[(2S,4S,5S)-l-((2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]2-[(methoxycarbonyl)amino]acetyI)-4,5-dimethylpynOlidin-2-yl]-l,lldihydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-9-yl}-lH-imidazol-2-yl)-2,3dimethylpyrrolidin-l-yl]-2-oxoethyl}carbamate. RT = 3.757 min (Gemini column, 2-95%
123
MeCN/H2O/0.1% TFA over 8 min). LCMS-ESI+: calc’d for C55H70N8O9: 986.53 (M+); Found: 987.88 (M+H+). ’H NMR (400 MHz, MethanokZ») δ 8.25 (s, 1H), 7.96 - 7.78 (m, 1H), 7.76 - 7.34 (m, 8H), 7.27 (s, 1H), 5.22 - 5.06 (m, 4H), 5.00 (t, 1H), 4.65 - 4.44 (m, 2H), 4.17-3.95 (m, 4H), 3.57 (s, 6H), 3.55-3.46 (m, 1H), 3.45-3.33 (m, 1H), 2.50- 1.97 (m, 9H), 1.55 (t, 2H), 1.41 (d, 3H), 1.37- 1.26 (m, 5H), 1.24-1.13 (m, 1H), 1.13 - 1.01 (m, 12H), 1.01-0.93 (m, 1H), 0.86 (d, 3H), 0.83-0.74 (m, 5H).
Example BA. Methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2[(2R,3R,5R)-5-(9-(2-[(2S,4S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyItetrahydro-2H-pyran-4yl]-2-[(methoxycarbonyl)amino]acetyl}-4,5-dimethylpyrrolidin-2-yl]-lH-imidazol-5-yl}l,ll-dihydroisochromeno[4',3’:6,7]naphtho[l,2-d]imidazol-2-yl)-2,3-dimethylpyrrolidin-lyl]-2-oxoethyl)carbamate. RT = 3.899 min (Gemini column, 2-95% MeCN/H2O/0.1% TFA over 8 min). LCMS-ESI+: calc’d for C55H70N8O9: 986.53 (M+); Found: 987.95 (M+H+). lH NMR (400 MHz, Methanol-d4) δ 8.43 - 8.28 (m, 1H), 8.05 - 7.88 (m, 1H), 7.88 - 7.42 (m, 6H), 7.39 - 7.25 (m, 1H), 5.31 - 5.01 (m, 4H), 4.70-4.55 (m, 1H), 4.46 - 4.18 (m, 2H), 4.18 - 4.05 (m,2H), 4.04 - 3.94 (m, 0H), 3.85-3.71 (m, 1H),3.65 (s, 5H), 3.52-3.38 (m, 1H), 2.63-1.98 (m, 9H), 1.75-1.50 (m, 3H), 1.47 (d,3H), 1.44-1.21 (m, 6H), 1.18-0.99 (m, 16H), 0.96 - 0.87 (m, 4H), 0.87 - 0.82 (m, 3H), 0.65 (d, J = 6.1 Hz, 1H).
Example BB. Methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2[(2R,3R,5R)-5-(5-{2-[(2S,4S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4yl]-2-[(methoxycarbonyl)amino]acetyl}-4,5-dimethylpyrrolidin-2-yl]-l,lld ihydroi sochromeno[4',3 ':6,7]naphtho[ 1,2-d]i midazol-9-yl} -1 H-i m idazol-2-yl)-2,3 dimethylpynOlidin-l-yl]-2-oxoethyl)carbamate. RT = 3.940 min (Gemini column, 2-95% MeCN/H2O/0.1% TFA over 8 min). LCMS-ESI+: calc’d for C55H70N8O9: 986.53 (M+); Found: 987.86 (M+H+). lH NMR (400 MHz, Methanol^) δ 8.23 - 8.03 (m, 1H), 7.79 7.64 (m, 2H), 7.59 - 7.20 (m, 6H), 7.15 - 7.02 (τη, 1 H), 5.04 - 4.87 (m, 3H), 4.83 (t, 1H), 4.43 - 4.27 (m, 1H), 4.27 -3.76 (m, 5H), 3.63 - 3.46 (m, 3H), 3.45 - 3.36 (m, 5H), 2.55 1.68 (m, 10H), 1.56-1.27 (m, 3H), 1.23 (d, J= 6.8 Hz, 1H), 1.20-1.11 (m, 5H), 1.08 (t,J= 7.1 Hz, 1 H), 1.04 - 0.73 (m, 18H), 0.73-0.51 (m, 5H), 0.01 (d, J = 6.0 Hz, 1 H).
Example BC. Methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyTan-4-yl]-2[(2R,3R,5R)-5-(5-{2-[(2R,4R,5R)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4y I]-2-[(methoxy carbony l)ami nojacety I} -4,5 -dimethy Ipyrroli d i n-2-y 1]-1,11dihydroisochromeno[4’,3':6,7]naphtho[l,2-d]imidazol-9-yl}-lH-imidazol-2-yl)-2,3dimethylpyrrolidin-l-yl]-2-oxoethyl)carbamate. RT = 4.076 min (Gemini column, 2-95%
124
MeCN/H2O/0.1% TFA over 8 min). LCMS-ESI+: calc’d for C55H70N8O9: 986.53 (M+); Found: 987.91 (M+H+). Ή NMR (400 MHz, Methanol-<A) δ 8.22 - 8.08 (m, 1H), 7.81 7.67 (m, 1H), 7.65 - 7.17 (m, 5H), 7.10 (s, 1H), 5.19 - 5.05 (m, 1H), 5.04-4.80 (m, 3H), 4.30-3.90 (m, 6H), 3.63-3.46 (m, 4H), 3.40 (s, 5H), 2.55 - 1.60 (m, 10H), 1.55 - 1.25 (m, 4H), 1.21 - 0.95 (m, 12H), 0.95 - 0.74 (m, 14H), 0.56 (d, 2H), 0.49 - 0.34 (m, 1H), 0.05 0.04 (m, 1H).
Example BD
methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2II-pyran-4-yl]-2-[(2S3S)-2-(5-{2[(2S,4S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydr(>-2H-pyran-4-yl]-2[(methoxycarbonyl)amino]acetyl}-4-methoxymethylpyrrolidin-2-yl]-l,lldihyd roisoch romeno]4',3’: 6,7] naphtho J1,2-d] imidazol-9-ylJ-l H-im idazol-2-y l)-5methylpyrrolidin-l-yl]-2-oxoethyl]carbamate.
Methyl {(lS)-l-[(2R,6R)-2,6-dimethy]tetrahydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(5-{2[(2S,4S)-l-((2S)-2-[(2R,6R)-2,6-dimethyItetrahydro-2H-pyran-4-yl]-2[(methoxycaibonyl)amino]acetyl}-4-methoxy methyl pyrrolidin-2-yl]-1,11dihydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-9-yl]-lH-imidazol-2-yl)-5methy1pyrrolidin-l-yl]-2-oxoethyl]carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)-5-methylpyrrolidme-2-carboxylic acid with (2S,5S)-l-(tertbutoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid and (2S,5S)-l-(tert-butoxycarbonyl)5-methy!pyTrolidine-2-carboxyIic acid with (2S,4S)-l-(tert-butoxycarbonyI)-4(methoxymethyl)pyrro1idine-2-carboxylic acid ’H NMR (400 MHz, dmso) δ 8.71 (s, 1H), 8.18 (m, 1H), 7.95 (m, 4H), 7.80-7.54 (m, 3H), 7.45 (m, 1H), 5.34-5.14 (m, 3H), 5.05 4.92 (m, 1H), 4.62 (s, 1H), 4.35 - 3.03 (m, 13H), 2.66 (s, 2H), 2.50 (m, 2H), 2.39 - 1.72 (m, 9H), 1.55 - 0.67 (m, 28H). MS (ESI) m!z 989.41 [M + H]+.
125
Example BE
O
O methy![(2S3S)-l-[(2S,5S>-2-(5-{2-[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydr(>2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyI}-5-methylpyrrolidin-2-ylj-l,4,5,U5 tetrahydroisochromeno[4*,3':6,7]naphtlio[l,2-d]imidazol-9-yI}-lH-imidazoI-2-yl)-5 methylpyrro!idin-l-yI]-3-methyI-l-oxopentan-2-yl}carbamate
Methyl{(2S,3S)-l-[(2S,5S)-2-(5-{2-[(2S,5S)-l-{(2S)-2’[(2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyI)amino]acetyl}-5-methylpyrrolidin2-yl]-l ,4,5,1 l-tetrahydroisochromeno[4’,3':6,7]naphtho[ l,2-d]imidazol-9-yl }-I H-imidazol-210 yl)-5-methylpyrrolidin-l-yl]-3-methyl-l-oxopentan-2-yl}carbamate was synthesized in a similar manner as example AA omitting the oxidatîon with MnO2. *H NMR (400 MHz, dmso) δ 7.84 (m, 4H), 7.62 (m, 2H), 7.53 (m, 1H), 7.25 (s, 1H), 5.15 (s, 2H), 5.02 - 4.88 (m, 2H), 4.62 (s, 2H), 4.14-3.24 (m, 16H), 3.06 (s, 2H), 2.88 (s, 2H), 2.21 (m, 8H), 1.82 (s, 2H), 1.67 (s, 1H), 1.44 (m, 8H), 1.30 - 0.95 (m, 8H), 0.91 (m, 3H), 0.78 (m, 5H), 0.64 (m, 3H).
MS (ESI) m/z 905.78 [M + H]+.
Example BF
126 methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(9-{2[(2S^S>-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2[(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin-2-yl]-lH-imidazol-5-yl}-l,4,5,l 1tetrahydroisochrOmeno[4'3':6,7]naphtho[l,2-d]imidazol-2-yl)-5-methylpyrrolidin-l-yl]2-oxoethy !} ca rba mate.
Methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(9-{2[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2[(methoxycarbonyl)aminojacetyl }-5-methylpyrrolidin-2-yl]-lH-imidazol-5-yl) -1,4,5,11tetrahydroisochromeno[4',3':6,7]naphtho[l,2-d]imidazol-2-yl)-5-methylpynOhdin-l-yl]’2oxoethyljcarbamate was synthesized in a stmîlar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3-methyIpentanoyl)-5-methy!pyrrolidine-2carboxylic acid with (2S,4S)-l-(tert-butoxycarbonyl)-5-methylpynOlidine-2-carboxylic acid and omitting the oxidation with MnOj. *H NMR (400 MHz, dmso) δ 8.05 — 7.75 (m, 4H), 7.73 - 7.47 (m, 3H), 7.31 (m, 1H), 5.15 (s, 2H), 4.96 (m, 2H), 4.61 (s, 2H), 4.15 -3.15 (m, 18H), 3.06 (s, 2H), 2.99 - 2.75 (m, 3H), 2.17 (m, 8H), 1.82 (m, 2H), 1.53 - 1.32 (m, 6H), 1.30 - 1.17 (m, 2H), 1.14 - 0.87 (m, 11H), 0.85 - 0.69 (m, 2H). MS (ESI) m!z 961.54 [M + H]+.
methyl{(2S3S)-l-i(2S4aS,6aS)-2-(5-{2-[(2S,5S>-l-{(2S)-2-[(2R,6R)-2,6dimethyltetrahydro-2II-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5methylpyrrolidin-2-yI]-l,ll-dihydroisochromeno[4'3f:<>,7]naphtho[l^-d]imidazol-9yl}-lH-imidazol-2-yl)hexahydrocyclopenta[b]pyrrol-l(2H)-ylI-3-methyl-l-oxopentan-2yljcarbamate
Methyl((2S,3S)-l-[(2S,3aS,6aS)-2-(5-{2-[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6d i methy It etrahydro-2H-pyran-4-y l]-2-[(methoxy carbony l)amino]acetyl} -5-methy Ipyrrol id in2-yl]-l, 11 -dihydroisochromeno[4',3’:6,7]naphtho[ 1,2-d]imidazol-9-yl }-l H-imidazol-2
127 yl)hexahydrocyclopenta[b]pyrrol-1 (2H)-yl]-3-methyl-1 -oxopentan-2-yl} carbamate was synthesîzed in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2(methoxycarbonylamino)-3-methylpentanoyl)-5-methylpyrrolidine-2-carboxylic acid with (2S,3aS,6aS)-l-((2S,3S)-2-(methoxycarbonylamino)-3methylpentanoyl)octahydrocyclopenta[b]pyrrole-2-carboxylic acid. ’H NMR (400 MHz, dmso) 5 8.62 (s, 1H), 8.18 (s, 2H), 7.90 - 7.64 (m, 3H), 7.63 - 7.39 (m, 2H), 5.24 (s, 2H), 5.13 (s, 1H), 5.02 (s, 1H), 4.69 (s, 2H), 4.13-3.85 (m, 3H), 3.47 (m, 10H), 2.87 (s, 2H), 2.42 (m, 2H), 2.10 (s, 5H), 1.75 (m, 5H), 1.51 (m, 7H), 1.31 -0.94 (m, 7H), 0.92 - 0.64 (m, 9H) MS (ESI) 929.46 [M + H] +.
Example BH methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetraIiydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(9-{2[(2S,4S)-l-{(2S)-2-[(2R,6R)-2,6-dimetliy!tetrahydro-2H-pyra!i-4-yl]-2|(methoxycarbonyl)amino]acetyl}-4-(methoxymethyl)pyrrolidin-2-yl|-lH-imidazol-5yl}-l,ll-dihydroisochroiiieno[4*,3’:6,7]naphtho[l,2-d]imidazol-2-yI)-5methylpyrroIidin-l-yl|-2-oxoethyl}carbamate.
Methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(9-{2[(2S,4S)-l-((2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2[(methoxycarbony 1 )amino]acetyl} -4-(methoxy m ethyl)pyrrol id in-2-y 1]-1 H-i mid azol-5-yl} 1,11 -di hy droisochromeno [4',3 6,7]naphtho[ 1,2-d]imi dazo!-2-yl)-5-methy 1 pyrrol i d i η-1 -y 1 ]-2oxoethyl)carbamate was synthesîzed in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3-methylpentanoyl)-5-methylpyrrolidine-2carboxylic acid with (2S,4S)-l-(tert-butoxycarbonyl)-4-(methoxymethyl)pyirolidine-2carboxylic acid. Ή NMR (400 MHz, cdcb) δ 8.69 (s, 1H), 8.28 - 8.09 (m, 2H), 8.00 - 7.75 (m, 3H), 7.62 m, 2H), 7.50 (m, 1H), 5.27 (s, 2H), 5.14 (, 1H), 4.73 (s, 1H), 4.33 - 3.25 (m, 20H), 2.66 (s, 1H), 2.58 -2.28 (m, 8H), 2.25- 1.79 (m, 4H), 1.65-0.66 (m, 21H). MS (ESI) m!z 989.65 [M + H]+.
128
Exemple BI
Methyl {(2S)-l-[(2S3aS,6aS}-2-(9-{2-I(2S,5S)-l-{(2S>-2-[(2R,6R>-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbony])ammo]acetyI}-5methy!pyrrolidin-2-yI]-lH-imidazoI-5-yl}-l,I l-dihydroisochromeno[4'3':6,7] naphthoIl,2-d]imidazol-2-y])hexahydrocyclopenta[b]pyrrol-l(2H)-yl]-3-inethyl-loxobu tan-2-y 1} ca rba mate.
Methyl {(2S)-l-[(2S,3aS,6aS)-2-(9-{2-[(2S,5S)-l-{(2S)-2-[(2R,6R>2,6dÎmethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyI)amino]acetyl}-5-methylpyrrolidin2-yl]-l H-imidazol-5-yl }-l, 11 -dihydroisochromeno[4',3':6,7] naphthof 1,2-d]imidazol-2y l)hexahydrocycl openta[b]pyrrol-1 (2H)-yl] -3-methyl-1 -oxobutan-2-yl} carbamate was synthesized in a similar manner as example AA substitutîng (2S,5S)-l-((2S,3S)-2(methoxycarbonylamino)-3-methylpentanoyl)-5-methylpyrrolîdîne-2-carboxylic acid with (2S,5S)-l-(tert-butoxycarbonyI)-5-methylpyrro!idine-2-carboxylic acid and (2S,5S)-l-(tertbutoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid with (2S,3aS,6aS)-l-((S)-2(methoxycarbonylamino)-3 methylbutanoyl)octahydrocyclopenta[b]pyrrole-2-carboxylic acid. ’H NMR (400 MHz, dmso) 5 8.67 (s, 1H), 7.72 (m, 8H), 5.33 - 5.17 (m, 3H), 5.00 (m, 1H), 4.77 (s, 1H), 4.62 (s, 1H), 4.17-3.86 (m, 5H), 3.49 (m, 10H),2.89(s, 1H), 2.56-1.70 (m, 7H), 1.47 (m, 6H), 1.30-0.97 (m, 6H), 0.90 (s, 4H), 0.81 (m, 8H). MS (ESI)m/z 915.37 [M + H]+.
129
Example BJ
O
methyl {(2S>l-I(2S,5S>-2-(9-{2-[(2S)-l-{(2S>-2-[(2R,6R)-2,6-dimethyltetrahydro-2Hpyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}pyrrolidin-2-yl]-lII-imidazol-5-yl}-l,l 1dihydroisochromeno[4’3'»6,7]naphtho[l,2-d]imidazoI-2-yl)-5-methylpyrrolidin-l-yl]-3 methyl-l -oxobutan-2-yl} ca rbamate.
Methyl {(2S)-l-[(2S,5S)-2-(9-{2-[(2S>l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}pyrrolidin-2-yl]-lH-imidazol-5-yl}-l,lldihydroisochromeno[4’,3':6,7]naphtho[l,2-d]îmidazol-2-yl)-5-methylpyrro!idin-l-yl]-3methyl-l-oxobutan-2-yl}carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3-methylpentanoyl)-5methylpyrrolidîne-2-carboxylic acid with (2S,5S)-l-(tert-butoxycarbonyl)-5methylpyrrolidine-2-carboxylic acid and (2S,5S)-l-(tert-butoxycarbonyl)-5methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-((S)-2-(methoxycarbonylamino)'3methylbutanoyl)-5-methylpynOlidine-2-carboxylic acid ’H NMR (400 MHz, dmso) δ 8.67 (s,
1H), 8.19 (m, 1H), 8.03 (m, 2H), 7.91 - 7.68 (m, 3H), 7.68-7.38 (m, 3H), 5.26 (s, 2H), 5.14 (m, 2H), 4.70 (s, 1H), 4.20 - 3.23 (m, 14H), 2.37 (s, 2H), 2.22 - 1.71 (m, 6H), 1.49 (m, 3H), 1.41 - 0.97 (m, 7H), 0.97-0.78 (m, 8H), 0.72 (m, 3H). MS (ESI) m/z 875.30 [M + H]+.
Example BK
130
Methyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(2S}-2-(5-{2-J(2S)-l{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yI]-2-|(methoxy carbony])amino]acetyl}pyrrolidin-2-yl]-l,ll-dihydroisochromenoi4',3':6,7] naphtho[l,2-d]imidazol-9-yl}-lH-imidazol-2-yI)pyrroIidin-l-yI]-2-oxoethyl}carbamate.
Methyl {(lS)-l-[(2R,6R)-2,6-dimethyItetrahydro-2H-pyran-4-yl]-2-[(2S>2-(5-{2[(2S)-l-((2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxy carbonyl)amino]acetyl}pynolidin-2-yl]-l,ll-dihydroisochromeno[4’,3':6,7] naphtho[l,2d]imidazol-9-y)}-lH-imidazol-2-yl)pyrrolidin-l-yl]-2-oxoethyl)carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2(methoxycarbonylamino)-3-methylpentanoyt)-5-methylpyrrolidine-2-carboxylic acid with (S)-l-(tert-butoxycarbonyl)pyrrolidine-2-carboxylic acid and (2S,5S)-l-(tertbutoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid with (S)-l-(tertbutoxycarbonyl)pyrroIidine-2-carboxylic acid. ’H NMR (400 MHz, dmso) δ 8.69 (s, IH), 8.19 (m, IH), 8.08 (s, IH), 7.86 (m, 3H), 7.73 (s, IH), 7.62 (m, IH), 7.47 (m, IH), 5.26 (s, 3H), 5.10 (m, IH), 4.12 - 3.24 (m, 18H), 2.37 (s, 2H), 2.31 - 1.89 (m, 8H). 1.31 (m. 6H), 1.06 (m, 7H), 0.96 - 0.76 (m, 8H) MS (ESI) m/z 931.86 [M + H]+.
Example BL
O
Methyl {(2S3S)-l-[(2S,5S)-2-(9-{2-I(2S,5S>-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro2H-pyran-4-yl]-2-[(methoxycarbonyI)amino]acetyl}-5-methylpyrrolidin-2-yl]-lHimidazoI-Syl}-l,ll-dihydroisochromeno[4’3':6,7] naphtho[l,2-dJimidazol-2-y!)-5methylpyrrolidin-l*yl]-3-methyl-l-oxopentan-2-yl}carbamate
Methyl {(2S,3S)-l-[(2S,5S)-2-(9-{2-[(2S,5S)-l-((2S)-2-[(2R,6R)-2,6dimethyltetrahydro-2H-pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5-methylpyrrolidin2-yl]-lH-imidazoI-5-yl}-l,ll-dihydroisochromeno[4',3':6,7] naphtho[l,2-d]imîdazol-2-yl)-5methylpynOlidin-l-yl]-3-methyl-l-oxopentan-2-yl}carbamate was synthesized in a similar manner as example AA substituting (2S,5S)-l-((2S,3S)-2-(methoxycarbonylamino)-3
131 methylpentanoyl)-5-methylpyrro!idine-2-carboxylic acid with (2S,5S)-1 -(tertbutoxycarbonyl)-5-methyIpyrroIidine-2-carboxylic acid and (2S,5S)-1 -(tertbutoxycarbonyl)-5-methylpyrrolidine-2-carboxylic acid with (2S,5S)-l-((2S,3S)-2(methoxycarbonylamino)-3-methylpentanoyl)-5-methylpynOlidine-2-carboxylic acid. 'H
NMR (400 MHz, dmso) δ 8.66 (s, IH), 8.18 (m, IH), 8.12-7.71 (m, 5H), 7.58 (m, 2H), 5.27 (s, 2H), 5.15 (m, IH), 5.06 - 4.93 (m, IH), 4.67 (m, 2H), 4.22-3.29 (m, 1 IH), 2.23 (m, 7H), 1.83 (s, 2H), 1.65 (s, IH), 1.47 (m, 8H), 1.29 - 0.98 (m, 7H), 0.95 - 0.70 (m, 8H), 0.66 (m, 3H). MS (ESI) mfz 903.87 [M + H]+.
Exemple BM
methyl {(1 S)-1 -[(2R.6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(2S>5S)-2-(5^2[(2S,5S)-1-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(rnethoxy carbonyOaminoIacetylJ-S-methylpyrrolidin^-yll-l.ll-dihydroisochromenoH'.S’ieJl naphtho[1,2-d]imidazol-9-yl}-1 H-imidazol-2-yl)-5-methylpyrn)IIdîn-1 -yl]-2-oxoethyl} carbamate
1. iodotrimethylsilane
2. ethyl carbamate
ethyl{(1S)-1-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(5-{2[(2S,5S)-1-{(2S)-2-[(2R,6R)-2.6-dimethyltetrahydro-2H-pyran-4-ylF2-[(ethoxy carbonyl)amino]acetyl}-5-methylpyrTOlidin-2-yl]-1,11-dihydroisochromeno[4*l3,:6,7] naphtho[1,2-d]imidazol-9-yl}-1 H-imidazo1-2-yl)-5-methylpyrrolidin-1 -yl]-2-oxoethyl} carbamate
Ethyl {(lS)-l-[(2K6R)-2,6-<limethyltetrahydn>-2H-pyran-4-yl]-2-|(2Sï5S)-2-(5-{2[(2S,5S)-l-{(2S)-2-[(2R,6R)-2,6-dimethyltetrahydro-2n-pyran-4-yl]-2[(ethoxycarbonyl)amino]acetyl}-5-methylpyrrolidin-2-yl]-l,lldihydroisochromenoI4'3,:6»7]naphtho[14-<i]iniidazoI-9-yl}-lH-imidazol-2-yl)-515 methylpyrroIidin-l-yl]-2-oxoethyIJ carbamate
Iodotrimethylsilane (1.14 ml, 8.03 mmol) is added to a solution of methyl {(1S)-1[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-[(2S,5S)-2-(5-{2-[(2S,5S)-l-{(2S)-2
132 [(2R56R)-2,6-dimethy!tetrahydro-2H’pyran-4-yl]-2-[(methoxycarbonyl)amino]acetyl}-5methylpyrro1idin-2-yl]-l,l l-dihydroîsochromeno[4',3':6,7]naphtho[l,2-d]imidazol-9-yl}-lHimidazol-2-yl)-5-methylpyrrolidin-l-yl]-2-oxoethyl}carbamate (770 mg, 0.8 mmol) in Dichloromethane (10 ml), and the mixture is then refluxed for 3 hours. After cooling to room température the mixture was concentrated, dissotved in ethyl acetate and extracted with 2x7 ml of IN hydrochloric acid solution. The aqueous phases are combined, cooled and then basified by addition of 5N sodium hydroxide. The basic aqueous phase is extracted with ethyl acetate 3 x 10 ml. The organic phases were combined, dried over Na^SCU and concentrated under vacuum, the product 667 mg (98.5%) was treated with sodium hydroxide (66.25 mg,
1.66 mmol) in Water (7 ml). The mixture was cooled in an ice bath and ethyl chloroformate (0.16 ml, 1.66 mmol) was added, the reaction mixture was stirred at 0°C for 30 min, extracted with 2 x 10 ml ethyl acetate. The combined organic layers were dried over Na^SCh and concentrated. The residue was purified by reverse phase HPLC (Gemini column, 10-46% MeCN/H2O/0.1% TFA). The desired fractions were combined, lyophilized to provide ethyl {(lS)-l-[(2R,6R)-2,6-dimethyltetrahydro-2H-pyran-4-yl]-2-I(2S,5S)-2-(5-{2-[(2S,5S)-l{(2 S)-2-[(2R,6R)-2,6-di methy ltetrahy dro-2H-pyran-4-yl ]-2-[(ethoxycarbony l)am inojacety 1} 5-methy!pyrrolidin-2-yl]-l,ll-dihydroisochromeno[4',3':6,7]naphtho[l,2-d]Îmidazol-9-yl}lH-îmidazol-2-yl)-5-methylpyrrolidin-l-yl]-2-oxoethyl}carbamate. lH NMR (400 MHz, dmso) δ 8,62 (s, 1H), 8.26-7.33 (m, 9H), 5.24 (s, 2H), 5.13 (s, 1H), 4.98 (s, 1H), 4.80-4.55 (m, 2H), 4.30 - 3.23 (m, 9H), 2.19 (m, 9H), 1.83 (s, 2H), 1.48 (m, 10H), 1.32-0.69 (m, 24H). MS (ESI) m!z 987.89 [M + H]+.
BIOLOGICAL ASSAYS
Effect of sérum proteins on replicon potency: Replicon assays are conducted in normal cell culture medium (DMEM + 10%FBS) supplemented with physiologie concentrations of human sérum albumin (40 mg/mL) or α-acid glycoprotein (1 mg/mL). ECsoS in the presence of human sérum proteins are compared to the ECso in normal medium to détermine the fold shift in potency.
MT-4 Cell Cytotoxicity: MT4 cells are treated with serial dilutions of compounds for a five day period. Cell viability is measured at the end of the treatment period using the Promega CellTiter-Glo assay and non-linear régression is performed to calculate CCso.
Compound Concentration Associated with Cells at ECso: Huh-luc cultures are incubated with compound at concentrations equal to ECso. At multiple time points (0 - 72
133 hours), cells are washed 2X with cold medium and extracted with 85% acetonitrile; a sample of the media at each time-point will also be extracted. Cell and media extracts are analyzed by LC/MS/MS to détermine the Molar concentration of compounds in each fraction. Représentative compounds of the disclosure hâve shown activity.
Solubility and Stability: Solubility is determined by taking an aliquot of 10 mM DMSO stock solution and preparing the compound at a final concentration of 100 μΜ in the test media solutions (PBS, pH 7.4 and 0.1 N HCl, pH 1.5) with a total DMSO concentration of 1%. The test media solutions are incubated at room température with shaking for 1 hr. The solutions will then be centrifüged and the recovered supematants are assayed on the HPLC/UV. Solubility will be calculated by comparing the amount ofcompound detected in the defined test solution compared to the amount detected in DMSO at the same concentration. Stability ofcompounds after an 1 hour incubation with PBS at 37°C will also be determined.
Stability in Cryopreserved Human, Dog, and Rat Hépatocytes: Each compound is incubated for up to 1 hour in hépatocyte suspensions (100 μΐ, 80,000°Cells per well) at 37°C. Cryopreserved hépatocytes are reconstituted in the serum-free incubation medium. The suspension is transferred into 96-well plates (50 pL/well). The compounds are diluted to 2 μΜ in incubation medium and then are added to hépatocyte suspensions to start the incubation. Samples are taken at 0, 10, 30 and 60 minutes after the start of incubation and reaction will be quenched with a mixture consisting of 0.3% formic acid in 90% acetonitrile/10% water. The concentration of the compound in each sample is analyzed using LC/MS/MS. The disappearance half-life of the compound in hépatocyte suspension is determined by fitting the concentration-time data with a monophasic exponential équation. The data will also be sealed up to represent intrinsic hepatic clearance and/or total hepatic clearance.
Stability in Hepatic S9 Fraction from Human, Dog, and Rat: Each compound is incubated for up to 1 hour in S9 suspension (500 μΐ, 3 mg protein/mL) at 37°C (n = 3). The compounds are added to the S9 suspension to start the incubation. Samples are taken at 0, 10, 30, and 60 minutes after the start of incubation. The concentration of the compound in each sample is analyzed using LC/MS/MS. The disappearance half-life of the compound in S9 suspension is determined by fitting the concentration-time data with a monophasic exponential équation.
134
Caco-2 Permeability: Compounds are assayed via a contract service (Absorption Systems, Exton, PA). Compounds are provided to the contracter in a blinded manner. Both forward (A-to-B) and reverse (B-to-A) permeability will be measured. Caco-2 monolayers are grown to confluence on collagen-coated, mîcroporous, polycarbonate membranes in 12well Costar TRANSWELL® plates. The compounds are dosed on the apical side for forward permeability (A-to-B), and are dosed on the basolateral side for reverse permeability (B-toA). The cells are incubated at 37°C with 5% CO2 in a humidified incubator. At the begtnning of incubation and at 1 hr and 2 hr after incubation, a 200-pL aliquot is taken from the receiver chamber and replaced with fresh assay buffer. The concentration of the compound in each sample is determined with LC/MS/MS. The apparent permeability, Papp, is calculated.
Plasma Protein Binding: Plasma protein binding is measured by equilibrium dialysis. Each compound is spiked into blank plasma at a final concentration of 2 μΜ. The spiked plasma and phosphate buffer is placed into opposite sides of the assembled dialysis cells, which will then be rotated slowly in a 37°C water bath. At the end of the incubation, the concentration of the compound in plasma and phosphate buffer is determined. The percent unbound is calculated using the following équation:
% Unbound = 100·
where Cf and Cb are free and bound concentrations determined as the post-dialysis buffer and plasma concentrations, respectively.
CYP450 Profiling: Each compound is incubated with each of 5 recombinant human CYP450 enzymes, including CYP1A2, CYP2C9, CYP3A4, CYP2D6 and CYP2C19 in the presence and absence of NADPH. Serial samples will be taken from the incubation mixture at the beginning of the incubation and at 5, 15,30,45 and 60 minutes after the start of the incubation. The concentration of the compound in the incubation mixture is determined by LC/MS/MS. The percentage ofthe compound remaining after incubation at each time point is calculated by comparing with the sampling at the start of incubation.
Stability in Rat, Dog, Monkey and Human Plasma: Compounds will be incubated for up to 2 hours in plasma (rat, dog, monkey, or human) at 37°C. Compounds are added to the plasma at final concentrations of 1 and 10 pg/mL. Aliquots are taken at 0, 5, 15,30,60,
135 and 120 minutes after adding the compound. Concentration of compounds and major métabolites at each time point are measured by LC/MS/MS.
Evaluation of cell-based anti-HCV activity: Antivira! potency (ECso) was determined using a Renilla luciferase (RLuc)-based HCV replicon reporter assay. To perform the assay for génotype 1 and 2a JFH-1, stable HCV la RLuc replicon cells (harboring a dicistronic génotype la H77 replicon that encodes a RLuc reporter), stable HCV • 1b RLuc replicon cells (harboring a dicistronic génotype lb Conl replicon that encodes a RLuc reporter), or stable HCV 2a JFH-1 Rluc replicon cells (harboring a dicistronic génotype 2a JFH-1 replicon that encodes a RLuc reporter, with L31 présent in NS5A) were dispensed into 384-well plates for ECso assays. To perform the assay for génotype 2a (with M31 présent in NS5A) or 2b, NS5A chimeric génotype 2a JFH-1 replicons that encodes a RLuc-Neo reporter and either génotype 2a J6 strain NS5A gene or génotype 2b MD2b-l strain NS5A gene (both with M31 présent) respecti vely, were either transiently transfected (t) into HuhLunet cells or were established as stably replicating replicon cells (s) is provided. Either cells were dispensed into 384-well plates for ECso assays. To perform the assay for génotype 3 and 4, NS5A chimeric génotype lb Conl replicons that encodes a Pi-RLuc reporter and either génotype 3a S52 strain NS5A gene or génotype 4a ED43 strain NS5A gene respectively, were transiently transfected (t) into Huh-Lunet cells, which were subsequently dispensed into 384-well plates. Compounds were dissolved in DMSO at a concentration of 10 mM and diluted in DMSO either manually or using an automated pipeting instrument. Serially 3-fold diluted compounds were either manually mixed with cell culture media and added to the seeded cells or directly added to the cells using an automated instrument. DMSO was used as a négative (solvent; no inhibition) control, and the protease inhibitor ITMN-191 was included at a concentration > 100 x ECso as a positive control. 72 hours later, cells were lysed and Renilla luciferase activity quantified as recommended by the manufacturer (PromegaMadison, WI). Non-linear régression was performed to calculate ECso values.
To détermine the antiviral potency (ECso) against résistance mutants, résistance mutations, including M28T, Q30R, Q30H, Q30E, L31M, Y93C, Y93H, and Y93N in génotype la NS5A, Y93H and L31V/Y93H in génotype lb NS5A, and Y93H for in génotype 3a NS5A, were introduced individually into either la Pi-Rluc or lb Pi-Rluc replicons by site directed mutagenesis. Replicon RNA of each résistant mutant was transiently transfected into Huh-7-derived cured-51 cells and antiviral potency was determined on these transfected cells as described above.
136
IV and PO Single Dose Pharmacokinetic Studies in SD Rats: The pharmacokinetics of selected compounds was characterized in male Sprague-Dawley (SD) rats (250-300g). In this study, two groups of naïve purebred SD rats (N=3 per group, fasted over night) received the selected compound either as an intravenous (IV) infusion (1 mg/kg over 30 minutes) via the jugular vein or by oral gavage (2 mg/kg). The intravenous (IV) dosing vehicle was 5% éthanol, 35% polyethylene glycol 400 (PEG 400) and 60% water pH 2.0. The oral dosing vehicle was 5% éthanol, 55% PEG 400 and 40% citrate buffer pH 2.2.
Serial blood samples (approximately 0.3 mL each) were collected from jugular vein or other suitable vein at specified time points. For the IV infusion group, the blood samples were collected predose and at 0.25,0.48, 0.58,0.75, 1.5,3,6,8, 12 and 24 hours after the start of infusion. For the oral group, the blood samples were collected predose and at 0.25, 0.50,1,2,4, 6, 8,12 and 24 hours after dosing. The blood samples were collected into Vacutainer’1' tubes containing EDTA-K3 as the anti-coagulant and were centrifuged at approximately 4°C to obtain plasma. The plasma samples were stored at -20°C until analysis by LC/MS/MS.
A bioanalytical method utilizing high performance liquid chromatography coupled to tandem mass spectrometry (LC/MS/MS) was developed for analysis of the selected compound in rat plasma. Détection was performed using selected reaction monitoring (SRM); Ions representing the precursor (M+H)+ species was selected in quadrupole 1 (Ql) and collided with argon gas in the collision cell (Q2) to generate spécifie product ion, which was subsequently monitored by quadrupole 3 (Q3). Standard curve and quality control samples were prepared in male rat plasma and processed in the same way as the test samples to generate quantitative data.
Pharmacokinetic parameters were generated using non-compartmental pharmacokinetic analysis (Phoenix WinNonlin, version 6.3). Values below the lower limit of quantification (LLOQ) were assïgned a value of zéro if predose and treated as missing thereafter. Area under the curve (AUC) was calculated using the linear trapézoïdal rule. The oral bioavailability (%F) was determined by comparison of the area under the curve (AUC) of the compound and/or a métabolite generated in plasma following oral administration to that generated following intravenous administration.
Data obtained in the above described assays for the compounds as described herein is shown in Table 1.
137
Compounds of this disclosure exhibit improved bi oavailability and/or activity towards certain HCV génotypes and/or résistant mutants thereof when compared to the comparative examples listed below. Comparative examples (Comp 1-14) as shown in Tables 2A and 2B below were prepared according to the synthetic protocols described herein using the appropriate starting materials.
138
BD AZ BA a BC AL AG AH AD AJ AN OV AP > /O AS AR AT AA AB > O AU AF AE 2 M p r
9600 0.031 190’0 | | 0.205 | 2.474 | 0.03 0.026 0.021 0.031 o o 00 0,021 o b £ | 0.049 o b LA 1 0.038 1 0.029 0.045 60’0 101*1 o b | 0.046 1 0.03 0.039 0.037 0.022 lb (nM)
90’0 o b kj 69 1'0 3.855 10.138 I 0.02 0.023 1 0.026 0.021 0.014 1 910Ό 600'0 4.444 0.012 1.518 6100 | | 0.026 | 8.983 | 1.518 9Ι0Ό 0.026 0.025 0.029 0.022 | 0.024 la (nM)
0.017 o ô 980’0 2.958 8.039 0.026 0.025 0.031 0.027 0.017 610’0 0.005 0.852 o b o 00 1.101 910’0 0.028 | 2.106 | 4.444 | ΙΪ0Ό 1100 1 9Ι0Ό 0.013 1 0.012 1 0.012 | 2a JFH (nM)
0.776 0.121 0.172 1.017 0.041 0.059 800’0 4.444 | 0.017 4.444 o b 0.015 | 5.137 | 900’0 0.028 | 0.003 | | 800’0 0.003 0.042 g, n*
800’0 0.015 0.016 0.026 0.014 0.024 0.020 o b 0.014 0.038
0.026 0.024 0.202 10.555 23.058 0.713 0.092 0.104 0.351 0.058 0.057 0.023 4 444 0.045 4.444 0.032 0.064 15.804 4.444 0.029 0.023 /—» NJ
0.072 0.025 0.231 10.35 23.3 980’0 0.137 0.154 0.074 0.028 0.035 o b 4.444 0.021 4.444 0.017 0,051 8.304 4,444 o o •U 0.025 610'0 0.029 9100 0.027 S 2»
0.029 0.013 0.108 2.537 6.235 o b NJ 00 o ô S 0.017 0.018 0.012 810’0 o b 0.935 0.018 1.188 o b NJ 0.024 4.191 1.888 o b o 00 900’0 500’0 o b 0.005 o b 'h' »
NJ -J 00 21.9 50.3 22.9 17.4 28.1 164 Rat %F
21.424 44.000 11.641 17.471 806’0 2.033 0.498 8990 1.332 0.059 08Γ0 ï 3 E _
0.208 0.160 la Q30R (nM)
2.106 1 2.700 1.213 1.784 0.074 P LA 0.145 0.082 0.312 0.042 0.097 la Q30E (nM)
0.088 o b LA 0.359 1 20.223 i 0,206 1 0.148 0.322 0.163 0.832 0,026 o NJ CN 00 0.230 la Y93H (nM)
0.104 0.059 0.496 29.566 4.054 4.303 4.651 9.034 1 0.337 1 0.325 0.570 1 0.418 1 1.634 680’0 i 668'0 1 1.121 o Ï2 » « 2
0.814 4.516 40.645 2.838 3.732 8.060 9.057 0.703 0.732 0.544 8.769 14.495 0.130 4.586 6.933 3a Y93H (»M)
Table 1
-U ο
w C0 to t W > > ra > Cfl » w Z B
2 r τ; $ «-rt < -< rtrt X >< O m ? r
ο ο ο r> o O o O o o O o o
b ο Χ^ o ό o o rtrt o o b o 9 rtrt
LA to ΧΟ to LU to to rtrt to rtrt x* M S *
LU X* οχ to OX 00 Xx 00 to 00 xo ***
ο b Ο ό Ο o ô o ‘o o o o o o o o o o b p O b o b M
tu rtrt to to LU to rtrt LA rtrt -U rtrt S *
Χχ οο xo ~O οχ ό to w Ό 00 Ό
Ο ο ο o O o o o o O o o O
ο ο o o ô b o o o o o O â Ίί?
to ο rtrt o rtrt l-rt rtrt rtrt rtrt rtrt to rtM < rt· »
lu Ό XO 00 LU to Ό **
Ο ο o O O o O o o O o
ο ό o ό o b b b b b b ® w
to LU to Ch o to LA Xx K LU S^j· 50
to LU Md OX 00 LU to LA Ch La Vi
ο ο O o o o O E
ό ο ô o ό o E,
to -U to to rtrt LU 2 -s·
** 00 XO XO LA o
o o o o O O /-V to
o o o o p o Ξ,
LU to to LU K to S'S'
LU ~j -J XO to XO
ρ ο ο ρ O a> o b o b o b o b o b o b O b o b o b K ω
χ^ to to to rtrt LA rtrt to Ch to M ®
LU -U LA LA xo LU OX LA 00 LA to
ο ο ο ο ο ’ο o b o o o o o o O b o b P o b o b o b -«-h X» 9 »
lu Q to o rtM rtrt rtrt rtrt to to rtrt 2c
ΧΟ δχ 00 LU U -J LA
LU to g5
to M rtrt “ rt*
xo £ ~
S* u» σ·
g- <
o<0
B LU **
5* O B sa
la Q30 (nM
M
Ο ο o o o o o O o o o O O
tO rtrt Ό LA ^rt LA rtrt o x^ LU o Ό Et Ό “
ΙΟ ο O o Xx LU 00 X* 00 Τ' LU LA
00 ο Ch o *“ X* o o x. LA LA
ο ο rtrt o o o o o o *<
la to -U o Md o 00 00 rtrt U E. b rt
LU La Ό xo LA 00 Ch 00 oo oo to to 2 LU fis
Lu o LU o Ch to LA LU LA LJ 'S Z
ο to 00 w o -U LA o to o ·<
ο to LA to LU X- b b LA LU to H. b <*>
X* Ch o Ox 0o oo LA rtrt Ch oo Ch LA te
-ο Lu Ch Ό oo U to Ch LU LA LU o œ
) s>° yy
cX1 <A r2·^ cA cA'
yz H >=z\
I-Zs^ “y
x JL l^jl fil Vi
L 1 [| J y! ΤΊ t
JL Ί Tk X ,o n P*
ΓιΓ IÎj ° XJ fil ° s
ιΎ |l J ί A
G A V^z-1 zx
aq zi Ad A? o<r 1 Λ A'Q
Co n o n o O o m M ·> {g
5 *o 2 “O 3 Ό 3 *o ^4 M ?
-U u> bO Λ
o o o o
o b o ô E, ** g cr
00 Os -J
P p P p S —
023 O NJ o LA LA o oo 2*
o o o o
b ô ô bJ ô o E ·Ί fc* s s s
oo Ό LA oo
0.026 O O Os O Ô OO GJ o b b3 - t* £ ® W ës “
o O
o ‘o GJ 3 *** K> g Ο σ
-U OO
O o o p
O o o SO 3$?
LA GJ bJ
o o o o
o b ô g “
u> LA Os ÏQ
Table 2A
ο /-—. -—1 yCr Q'r cç5 ,çy . -Ai /
Λ A ΙΙΊ îS Cl
d Tl II Tl 2
lîT n R*
ÎJO jlT° U Γχ C n n
u>9 ~L yO Ar' T zx À y?
Ό' ’ti AO 0
n n n n n M M
3 *T3 B Ό B Ό B Ό B •o 5? K ? B
Ό 00 -J Os Lft Ό ifi
0.034 0.04 0.028 O o <y» 0.029 'Ô' M z =
o p o p p s* —
b K» oo o tsj 00 O oo O M -U o N» K> M **
o o o o O
‘o o ô b b a b
o w Lrt K> U) S *“
o o o o
’o Lu LA .012 .012 3, σ\ κ> Z3 B
o o o o “s
ô b b O gS s
LA -J N> -U
o o o o o
LU LU .088 .038 .054 |S s
o o o o o
.08 .07 .03 .07 .04 EL w 5* ^3
Lu LA Lh
θ' J Y a y Q , o/y xV”
T M 5r' Y ï 7-\
«O 33 J x-zJ
A A. fil A. A Vi
Tl LA 11 c
JL^,o iiT AJ AJ cr n c
JJ ΪΊ ilT 3
Œ I1T LA. X-ÿÀ LA-1
JX' rA-Q 0-j Ati 5 Y' ;?ι AQ A s-Tw ”50
n o ΓΪ o r> o Ci o n o m M
2 T3 3 Ό 3 •o 3 Ό 3 •o Z e « ¢0 3
-U U) to o fb
0.025 o ô x» oo o o to to O X. σ> o o to Xi s s* M M C
o O P o P IT
O to <Z> O to IZI o Ui to o Ui o LA 5* M
o o P O p □ h
o o U) o o o -U to ** Ξ ·*· e
o o o o Ξ «-
.053 o bO to o IO tO o to o to Xi 5 Cb 5*^-. to »
0.036 o ô UJ UJ S* go K> cr
o ô -U oo o ô -J o to o LU o o Cb to 20 to σ·
O o o o O Xi O O Έ* ω ¢5
*-4 o -J to M
. -Λ i 1 X ^5-¾ 1 x i Q \ c/ .Ar V ÇC. Çb \A. AO J. Y A 1 A Q ^o> % I-V Çb ùfr' 1 o / .ΛΥ ï y£\ Ab Structure
n a n n n M
o o o o o
B B B B B © B
•a o Ό Ό •o p ·©
GH -U GA bJ rt
o o o o o <—» *
ô ô o ô ô S
bJ bJ bJ
4x N> 05
22 to p 46.6 50 50 Rat %F
0.971 S-5E= = ΐ
o o o o ο O
ô ô o o E, y ~
GA GA GH 05
bJ b-> « SC
o to o o GH o ω o© & SS -
50 GA bJ ·-—
--J bJ 05 50 « PI
0.46 o <o -J K> GH GA o < H. Ό 2 £69
o> 00 -J * K
bJ 'm'
o 05 O -U â v »* Z £ *
bJ -J “ Z
o o oo 'b
gh 05 50 O ô -J ® 1© ω 2 3 »
Ό GH i* GA
Table 2B
J. 'Γ° ÀC Ο 'X r~\ a y X LO T
ί^^ιΐ ï ΙίΊ JL JL Cfl
Tl C jL CA OL OL 2
ΙΪΊ ° JU° JO fï c
JO ° JO n
VU OL_
Ot <O U?? A ,Λ>
•ή. O 1 Q’ ‘Ό ,Ar ' AQ’
n n n O o w
B Ό o B •q o B Ό o B •Q o B T3 Z g ? £
O *o 00 -J Os Έ. ST
K> O O 's'1
00 p ko c Ό o 4b 3^
P Ln o
-~J b> •q 5·
p θ' so E» —
o w -U 2 g < θ’
o o O ο O
KJ bo o o 4b 3 £ »
VJ o bJ - »
o o O ο O
AJ ko --J o ** Lu 2 g »
o Ό pi
LU o -o
4b Ό Lrt ko o 4b 4b b» S \Λ 2 S 89
3.73 P LA 4b 1« -J o ·< » \o *2y ®
Ό 00 o O 4? z
21.8 4b O ko O bJ t-Λ bJ o *< 3 V© w 2 3 »
t. A V M v Q , crr P
A ά Çl JZ C
rV JL o U Πc *1 n>
P 'â>- Àb 05 Zï rp Ï-Ck AO
n o n o o o Q o M
2 o 5 XJ 5 xj 5 XJ Z » P-t
-U U) to rT
U lu Ό -J I£ iî ***
6.882 LA Ch -U u> 2? •Ί £.
0,130 o o XO -J = 5 C - 2 σ
1.195 to QO U) ο Ό s-os “ SO
p U> Ό oO g p ~ F5
3.664 o -J -U tO U -U O * ® xo μ2 <*· » ~ e
1.746 N N> 00 u» Ό XD 's 5 \Λ 2 S »
-U lu -J O »< s \O tü g S »
-U σχ
147

Claims (53)

  1. Claims
    1. A compound of formula 0):
    Ει*-ν’* -C(=O}-P’* -Wu -Plb-C(=O)-Vlb-Elb (I) wherein:
    Wu is and W11 is optionally substituted with one or more halo, alkyl, haloalkyl, optionally substituted aryl, optionally substituted heterocycle, or cyano;
    Y5 is -O-CH2-, -CH2-O-, -O-C(=O)-, or -C(=O)-O-;
    X5 is -CH2-CH2-, or -CH-CH-;
    P1* and P’b are each independently:
    Vu and Vlb are each independently:
    148 provided that at least one ofV1* and VIbis ^.or**^ ;
    E*1 and Elb are each independently -N(H)(alkoxycarbonyl), -N(H)(cycloalkylcarbonyl), or -N(H)(cycloalky!oxycarbonyl); or E11-''/'1 taken together are R91; or E,b-V,b taken together are R9b; and
    R91 and R9b are each independently:
    Ά »-r° ° or O ;
    or a pharmaceutically acceptable sait or prodrug thereof.
  2. 2. The compound of any preceding claim, which has the formula:
    (A1)
    149 wherein the imidazole ring shown in formula Al, A2, A3, and A4 is optionally substituted with one or more halo, haloalkyl, cyano, or alkyl;
    or a pharmaceutically acceptable sait or prodrug thereof.
  3. 3. The compound of any preceding claim, which has the formula:
    (A2) wherein the imidazole ring shown in formula A2 and A4 is optionally substituted with one or more halo, haloalkyl, cyano, or alkyl;
    or a pharmaceutically acceptable sait or prodrug thereof.
    150
  4. 4, The compound of any preceding claim, wherein Pu and P,b are each independently:
  5. 5. The compound of any preceding claim, wherein Vu and V,b are each independently:
  6. 6. The compound of claim 1, wherein:
    Wla is and W1* is optionally substituted with one or more halo, alkyl, haloalkyl, or cyano;
    YJ is -O-CH2-, or-CH2-O-;
    X5 is -CH2-CH2-, or -CH=CH-;
    P1’ and P,b are each independently:
    151
    V11 and VIb are each independently:
    provided that at least one of V1* and Vlb is
    E1* and Elb are each independently -N(H)(alkoxycarbonyl), -N(H)(cyc!oalkylcarbonyl), or -N(H)(cycloalkyloxycarbonyl); or E’*-Vl* taken together are R9*; or EIb-VIb taken together are R96; and
    R9* and R9b are each independently:
    ΖγΟ /J3 */ -y'O or O ;
    or a pharmaceutically acceptable sait or prodrug thereof. 7
  7. 7. The compound of any one of daims 1 -6, wherein one of Vla and Vlb is:
    **^· ^,01^ and the other ofV1* and Vlb is
    152
  8. 8.
    The compound of any one of claims l-6, wherein one of V1* and VIb is:
    and the other of V11 and Vlb is
  9. 9.
    The compound of any one of claims 1-6, wherein one of V11 and V,b is:
    5:
    , and the other of V1· and V,b is
  10. 10.
  11. 11.
    The compound of any one of claims 1-6, wherein one of V11 and Vlb is:
  12. 12.
    ***»· and the other of V11 and Vlb is
    The compound of any one of claims 1-6, wherein one of Vu and VIb is:
    I °x t and the other of V1* and Vlb is
    153
  13. 13.
    The compound of any one of claims 1-6, wherein one of V1* and V,b is:
    or
  14. 14.
  15. 15. The compound of any one of claims 1-6, wherein one of V1’ and V’b is:
  16. 16. The compound of any preceding claim, wherein P11 and P,b are each independently:
  17. 17. The compound of any one of claims 1-6, wherein one of V11 and Vlb is:
    154
  18. 18. The compound of any one of daims 1-6, wherein both of V1* and Vlb are:
  19. 19. The compound of any one of daims 1-6, wherein one of Vla and Vlb is:
  20. 20. The compound of any one of daims 1-6, wherein both of V1* and Vlb are:
  21. 21. The compound of any one of daims 1-6, wherein one of V1* and V,b is:
    , provided that bond (a) is connected to E1* or E’b and bond (b) is connected to the
    -C(=O)- group of formula (1) or (Al, A2, A3, or A4).
  22. 22. The compound of any one of daims 1-6, wherein one of VIa and V,b is:
    H , provided that bond (a) is connected to E1* or Elb and bond (b) is connected to the -C(=O)- group of formula (1) or (Al, A2, A3, or A4).
    155
  23. 23. The compound of any one of daims 1-6, wherein one of V1* and Vlb is:
    , provided that bond (a) is connected to E1* or E,b and bond (b) is connected to the
    -C(=O)- group of formula (1) or (Al, A2, A3, or A4).
  24. 24. The compound of any one of daims 1-6, wherein one of V’1 and VIb is: provided that bond (a) is connected to Eu or Elb and bond (b) is connected to the
    -C(=O)- group of formula (1) or (Al, A2, A3, or A4).
  25. 25. The compound of any one of daims 1-24, wherein one of P1* and Plb is:
  26. 26. The compound of any one of daims 1-24, wherein one of P11 and P,b is:
  27. 27. The compound of any one of daims 1-24, wherein both of P1* and P,b are:
    156
  28. 28. The compound of any one of claims 1-6, wherein -V^-C^O)-?1’- and -Plb-C(=O)-V,bare each independently:
    157 provided that at least one of V1* and Vlb is
    158
  29. 29. The compound of any one of claims 1-6, wherein -Vlt-C(=O)-P11- and -Plb-C(=O)-V,bare each independently:
  30. 30. The compound of any one of claims 1 -6, wherein one of-Vh-C(=O)-P’·- and
    159
    160
  31. 31. The compound of any one of cîaims 1-6, wherein one of-V^-C^CO-P1*- and
    -PIb-C(=O)-Vlb- is:
    and the other of-V'^C^O)-?'*- and -Plb-C(=O)-Vlb- is:
  32. 32. The compound of any one of daims 1-6, wherein both of -V’*-C(=O)-PIa- and
    -Plb-C(=O)-V,b- are:
    V
    VY°
  33. 33. The compound of any preceding daim, wherein at least one of EIa and Elb is -N(H)(alkoxycarbonyl).
  34. 34. The compound of any preceding claim, wherein both of E1* and Elb are -N(H)(alkoxycarbonyl).
    161
  35. 35. The compound of any preceding claim, wherein at least one of E1* and E’b is -N(H)C(=O)OMe.
  36. 36. The compound of any preceding claim, wherein both of E1* and E,b are -N(H)C(=O)OMe.
  37. 37. A compound selected from the group consisting of:
    162 ο
    163
    V»**’
    164 or a pharmaceutically acceptable sait or prodrug thereof.
    165 ο
    / ο.
    166 /
    167 p·*·'
    H
    168 or a pharmaceutically acceptable sait or prodrug thereof.
  38. 39. A compound having the formula:
    169
  39. 41. A pharmaceutical composition comprising the compound as described in any preceding claim or a pharmaceutically acceptable sait, or prodrug thereof; and at least one pharmaceutically acceptable carrier.
  40. 42. The pharmaceutical composition of claim 41 for use in treating disorders hepatitis C (HCV).
  41. 43. The pharmaceutical composition of claim 41, further comprising at least one additional therapeutic agent for treating HCV.
  42. 44. The pharmaceutical composition of claim 43, wherein said additional therapeutic agent is selected from ribavirin, an NS3 protease inhibitor, a nucleoside or nucléotide inhibitor of HCV NS5B polymerase, an alpha-glucosidase 1 inhibitor, a hepatoprotectant, a non-nucleoside inhibitor of HCV polymerase, or combinations thereof.
  43. 45. The pharmaceutical composition according to claim 41, further comprising a nucleoside or nucléotide inhibitor of HCV NS5B polymerase.
  44. 46. The pharmaceutical composition according to daim 45, wherein said nucleoside or nucléotide inhibitor of HCVNS5B polymerase is selected from ribavirin, viramidine, levovirin, a L-nucleoside, or isatoribine.
    170
  45. 47. A pharmaceutical composition comprising a compound as in daims 1-41, at least one nucleoside or nucléotide inhibitor of HCV NS5B polymerase, and at least one pharmaceutically acceptable carrier.
  46. 48. The pharmaceutical composition according to claim 47, further comprising an interferon, a pegylated interferon, ribavirin or combinations thereof.
  47. 49. The pharmaceutical composition of any one of claims 47-48, wherein the nucleoside or nucléotide inhibitor of HCV NS5B polymerase is sofosbuvir.
  48. 50. A pharmaceutical composition comprising a compound as in claims 1-41, at least one NS3 protease inhibitor, and at least one pharmaceutically acceptable carrier.
  49. 51. The pharmaceutical composition according to claim 50, further comprising sofosbuvir.
  50. 52. A method of treating hepatitis C, said method comprising administering to a human patient a pharmaceutical composition which comprises a therapeutically effective amount of the compound as described in any of claims 1-41 or a pharmaceutically acceptable sait, or prodrug thereof.
  51. 53. The method according to claim 52, further comprising administering to the patient an interferon or pegylated interferon.
  52. 54. The method of any one of claims 52-53, further comprising administering to the patient ribavirin.
  53. 55. A compound as described in any one of claims 1 -41 or a pharmaceutically acceptable sait, or prodrug thereof for use in medical therapy.
OA1201500241 2012-12-21 2013-12-19 Antiviral compounds. OA17337A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US61/745,452 2012-12-21
US13/830,346 2013-03-14

Publications (1)

Publication Number Publication Date
OA17337A true OA17337A (en) 2016-05-25

Family

ID=

Similar Documents

Publication Publication Date Title
AU2013361195B2 (en) Antiviral compounds
US10800789B2 (en) Antiviral compounds
AU2015274863B2 (en) Antiviral compounds
OA17337A (en) Antiviral compounds.
OA17167A (en) Condensed imidazolylimidazoles as antiviral compounds.