USOO894.0718B2

(12) United States Patent (10) Patent No.: US 8,940,718 B2 Bacon et al. (45) Date of Patent: *Jan. 27, 2015

(54) ANTIVIRAL COMPOUNDS (56) References Cited (71) Applicant: Gilead Sciences, Inc., Foster City, CA U.S. PATENT DOCUMENTS (US) 8,575,135 B2 * 1 1/2013 Bacon et al...... 514.92 2013/0309196 A1 11/2013 Bacon et al. (72) Inventors: Elizabeth M. Bacon, Burlingame, CA 2014/O112885 A1 4/2014 Bacon et al. (US); Jeromy J. Cottell, Redwood City, CA (US); Ashley Anne Katana, San FOREIGN PATENT DOCUMENTS Mateo, CA (US); Darryl Kato, San Francisco, CA (US); Evan S. WO WO 2008/021927 2, 2008 Krygowski, Washington, DC (US); John WO WO 2008/021928 2, 2008 WO WO 2008/021936 2, 2008 O. Link, San Francisco, CA (US); WO WO 2009/102318 8, 2009 James Taylor, San Mateo, CA (US); WO WO 2009/102325 8, 2009 Chinh Viet Tran, San Mateo, CA (US); WO WO 2009/102633 8, 2009 Teresa Alejandra Trejo Martin, Union WO WO 2010/062821 6, 2010 WO WO 2010/065681 6, 2010 City, CA (US); Zheng-Yu Yang, Palo WO WO 201O/O96777 8, 2010 Alto, CA (US); Sheila Zipfel, San WO WO 2010/099527 9, 2010 Mateo, CA (US) WO WO 2010, 132538 11, 2010 WO WO 2010, 1326O1 11, 2010 (73) Assignee: Gilead Pharmasset LLC, Foster City, WO WO 2010, 138368 12/2010 WO WO 2011/028596 3, 2011 CA (US) WO WO 2011/066241 6, 2011 WO WO 2011 O75439 6, 2011 (*) Notice: Subject to any disclaimer, the term of this WO WO 2011/087740 T 2011 patent is extended or adjusted under 35 WO WO 2011/112429 9, 2011 U.S.C. 154(b) by 0 days. WO WO 2011, 146401 11, 2011 WO WO 2012/027712 3, 2012 This patent is Subject to a terminal dis WO WO 2012/048421 4/2012 claimer. WO WO 2012/068234 5, 2012 WO WO 2012,087.976 6, 2012 (21) Appl. No.: 13/679,874 OTHER PUBLICATIONS (22) Filed: Nov. 16, 2012 U.S. Appl. No. 13/679,862, filed Nov. 16, 2012, Bacon et al. International Search Report and Written Opinion for PCT/US2012/ (65) Prior Publication Data 065681 dated Jan. 25, 2013. International Search Report and Written Opinion for PCT/US2011/ US 2013/O156732 A1 Jun. 20, 2013 060966 dated Sep. 19, 2012. Lam et al., “Genotype and Subtype Profiling of PSI-7977 as a Nucleotide Inhibitor of .” Antimicrobial Agents and Related U.S. Application Data Chemotherapy, Jun. 2012, vol. 56, No. 6, pp. 3359-3368. STN Registry No. 1 190307-88-0, “.” Retrieved from STN (60) Provisional application No. 61/560,654, filed on Nov. Registry File No. Oct 25, 2013 (1 page). 16, 2011. U.S. Office Action, dated Oct. 29, 2013, in corresponding U.S. Appl. No. 13/679,862 (13 pages). Int. C. U.S. Appl. No. 14/261.325, filed Apr. 24, 2014, Bacon, et al. (51) U.S. Appl. No. 14/316,573, filed Jun. 26, 2014, Cottell, et al. AOIN 43/16 (2006.01) U.S. Office Action, datedMar. 26, 2014, incorresponding U.S. Appl. A6 IK3I/35 (2006.01) No. 14/049,133 (8 pages). AOIN33/18 (2006.01) AOIN33/24 (2006.01) * cited by examiner A6 IK3I/04 (2006.01) CO7D 207/216 (2006.01) Primary Examiner — Jeffrey S. Lundgren A6 IK3I/488 (2006.01) Assistant Examiner — Chris Simmons A6 IK 45/06 (2006.01) (74) Attorney, Agent, or Firm — Sheppard Mullin Richter & C07D 49 I/052 (2006.01) Hampton LLP (52) U.S. C. CPC ...... A61K 45/06 (2013.01); A61 K3I/4188 (57) ABSTRACT (2013.01); C07D491/052 (2013.01); Y10S The disclosure is related to anti-viral compounds, composi 514/894 (2013.01) tions containing such compounds, and therapeutic methods USPC ...... 514/92: 514/894: 514/741; 514/456: that include the administration of such compounds, as well as 548/571 to processes and intermediates useful for preparing Such com (58) Field of Classification Search pounds. USPC ...... 514/894, 741, 453: 548/571 See application file for complete search history. 28 Claims, No Drawings US 8,940,718 B2 1. 2 ANTIVIRAL COMPOUNDS -continued CROSS REFERENCE TO RELATED APPLICATIONS This application claims the benefit under 35 U.S.C. S 119 (e) of U.S. Provisional Application No. 61/560,654 filed on Nov. 16, 2011, which is hereby incorporated by reference in its entirety. 10 BACKGROUND P' is selected from: Hepatitis C is recognized as a chronic viral disease of the liver which is characterized by liver disease. Although drugs N targeting the liver are in wide use and have shown effective 15 N ness, toxicity and other side effects have limited their useful ness Inhibitors of hepatitis C virus (HCV) are useful to limit the establishment and progression of by HCV as well as in diagnostic assays for HCV. There is a need for new HCV therapeutic agents. In par F O ticular, there is a need for HCV therapeutic agents that have N 19 broad activity against HCV genotypes (e.g. genotypes 1a, 1b, F 2a, 3a, 4a). There is also a particular need for agents that are less Susceptible to viral resistance. Resistance mutations to N inhibitors have been described for HCV NS5A for genotypes 25 1a and 1b in Antimicrobial Agents and Chemotherapy, Sep 9, N tember 2010, Volume 54, p. 3641-3650. Y. O ra s SUMMARY 30 P' is selected from: In one embodiment the disclosure provides a compound of the disclosure which is a compound of formula (I): Ela-Vila C(—O) Pia Wa P 1 C(—O) v1b-E1b (I) wherein: 35 y 4. W is N N Ys 40 N \ { Yi X5 F O 45 N 1. O F and W' is optionally substituted with one or more groups independently selected from halo, alkyl, haloalkyl, or cyano; N Y is —O CH , or —CH. O. : X is —CH, N CH- or -CH=CH-; E" is N(H)(alkoxycarbonyl), N(H)(cycloalkylcarbo 50 y O s s nyl) or N(H)(cycloalkyloxycarbonyl); or E'-V' taken together are R': E' is N(H)(alkoxycarbonyl), N(H)(cycloalkylcarbo nyl) or N(H)(cycloalkyloxycarbonyl); or E'-V' taken and together are R': 55 R" and Rare each independently: V" and V' are each independently selected from:

60 O H1 N H1 N

O O 65 \, , , or a pharmaceutically acceptable salt or thereof. US 8,940,718 B2 3 4 The disclosure also provides isotopically enriched com In another aspect the disclosure provides a method of pounds that are compounds of the disclosure that comprise an inhibiting HCV activity in a sample comprising treating the enriched isotope at one or more positions in the compound. sample with a compound of the disclosure. The present disclosure also provides a pharmaceutical Compounds of formula (I) have been found to possess composition comprising a compound of the disclosure or a useful activity against HCV genotypes. Additionally certain pharmaceutically acceptable salt or prodrug thereof and at compounds of formula (I) have significant potency against least one pharmaceutically acceptable carrier. resistant variants in GT1. The present disclosure also provides a pharmaceutical Accordingly, certain compounds of formula (I) possess composition for use in treating hepatits C(HCV). In one beneficial pharmacological properties that make them well embodiment the composition comprises at least one addi 10 suited to fulfill the current need for HCV agents with such tional therapeutic agent for treating HCV. In one embodi beneficial properties. ment, the therapeutic agent is selected from , an NS3 In one embodiment the disclosure provides a compound protease inhibitor, a nucleoside or nucleotide inhibitor of having improved inhibitory or pharmacokinetic properties, HCV NS5B polymerase, an alpha-glucosidase 1 inhibitor, a 15 including enhanced activity against development of viral hepatoprotectant, a non-nucleoside inhibitor of HCV poly resistance, improved oral bioavailability, greater potency (for merase, or combinations thereof. In one embodiment, com example, in inhibiting HCV activity) or extended effective position further comprises a nucleoside or nucleotide inhibi half-life in vivo. Certain compounds of the disclosure may tor of HCV NS5B polymerase. In one embodiment, the have fewer side effects, less complicated dosing schedules, or nucleoside or nucleotide inhibitor of HCV NS5B polymerase be orally active. is selected from ribavirin, viramidine, levovirin, a L-nucleo side, or isatoribine. DETAILED DESCRIPTION In one embodiment is provided a pharmaceutical compo sition comprising a compound as described herein and at least Reference will now be made in detail to certain embodi one nucleoside or nucleotide inhibitor of HCV NS5B poly 25 ments of the disclosure, examples of which are illustrated in merase, and at least one pharmaceutically acceptable carrier. the accompanying structures and formulas. While the disclo In one embodiment, the composition further comprises an sure will be described in conjunction with the enumerated , a pegylated interferon, ribavirin or combinations embodiments, it will be understood that they are not intended thereof. In one embodiment, the compound is the compound to limit the disclosure to those embodiments. On the contrary, exemplified in Example PY. In one embodiment the nucleo 30 the disclosure is intended to cover all alternatives, modifica side or nucleotide inhibitor of HCV NS5B polymerase is tions, and equivalents, which may be included within the Sofosbuvir. scope of the present disclosure as defined by the embodi The present disclosure also provides a pharmaceutical mentS. composition further comprising an interferon or pegylated Compounds interferon. 35 The present disclosure also provides a pharmaceutical The compounds of the disclosure exclude compounds composition further comprising a nucleoside analog. heretofore known. However, it is within the disclosure to use The present disclosure also provides for a pharmaceutical compounds that previously were not known to have antiviral composition wherein said is selected properties for antiviral purposes (e.g. to produce an anti-viral from ribavirin, Viramidine, levovirin, an L-nucleoside, and 40 effect in an animal). With respect to the United States, the isatoribine and said interferon is C-interferon or pegylated compounds or compositions herein exclude compounds that C-interferon. are anticipated under 35 USC S102 or that are obvious under The present disclosure also provides for a method of treat 35 USC S103. ing hepatitis C, said method comprising administering to a Whenever a compound described herein is substituted with human patient a pharmaceutical composition which com 45 more than one of the same designated group, e.g., "R" or prises a therapeutically effective amount of a compound of “A”, then it will be understood that the groups may be the the disclosure. same or different, i.e., each group is independently selected. The present disclosure also provides a method of inhibiting “Absent’ Some groups are defined such that they can be HCV, comprising administering to a mammal afflicted with a absent. When a group is absent it becomes a bond connector. condition associated with HCV activity, an amount of a com 50 The two groups that would otherwise be connected to that pound of the disclosure, effective to inhibit HCV. absent group are connected to each other through a bond. The present disclosure also provides a compound of the The “P” groups (e.g. P' and P') defined for formula (I) disclosure for use in medical therapy (e.g. for use in inhibiting herein have one bond to a —C(=O)—of formula (I) and one HCV activity or treating a condition associated with HCV bond to a W' group. It is to be understood that a nitrogen of activity), as well as the use of a compound of the disclosure 55 for the manufacture of a medicament useful for inhibiting the P group is connected to the —C(=O)—group of formula HCV or the treatment of a condition associated with HCV (I) and that a carbon of the P group is connected to the W' activity in a mammal. group. The present disclosure also provides synthetic processes and novel intermediates disclosed herein which are useful for 60 preparing compounds of the disclosure. Some of the com YS H pounds of the disclosure are useful to prepare other com N N N pounds of the disclosure. s \ ( In another aspect the disclosure provides a compound of V X5 N the disclosure, or a pharmaceutically acceptable salt or pro 65 H drug thereof, for use in the prophylactic or therapeutic treat ment of hepatitis C or a hepatitis C associated disorder. US 8,940,718 B2 5 In the W" group aYgroup is present. When that Ygroup is defined as —O—CH2—, or—CH2—O— group, those Y groups have a directionality. The Y group is connected to the W' group in the same left to right directionality that each is drawn. So for example, when Y is —O—CH2—, the directly 5 following structure is intended: Alkenyl is C-Cs hydrocarbon containing normal, sec O ondary, tertiary or cyclic carbonatoms with at least one site of 10 unsaturation, i.e. a carbon-carbon, sp' double bond. H Examples include, but are not limited to, ethylene or vinyl f (—CH=CH-), allyl ( CH-CH=CH), cyclopentenyl N (—CH), and 5-hexenyl ( CHCHCHCH-CH=CH-). - H N Alkynyl is C-Cs hydrocarbon containing normal, sec 15 ondary, tertiary or cyclic carbonatoms with at least one site of unsaturation, i.e. a carbon-carbon, sp triple bond. Examples For example, when Y is —CH. O. , the directly fol include, but are not limited to, acetylenic ( C=CH) and lowing structure is intended: propargyl ( CH-C=CH). Alkylene' refers to a saturated, branched or straight chain

O or cyclic hydrocarbon radical of 1-18 carbon atoms, and having two monovalent radical centers derived by the H removal of two hydrogen atoms from the same or two differ ent carbon atoms of a parent alkane Typical alkylene radicals N d include, but are not limited to, methylene (-CH2—) 1.2- 25 ethyl (-CHCH ), 1,3-propyl (—CH2CHCH ), 1,4- - VH butyl ( CHCH2CHCH ), and the like. Alkenylene' refers to an unsaturated, branched or straight In the structure I, the W' group has a left-to-right direc chain or cyclic hydrocarbon radical of 2-18 carbonatoms, and tionality as depicted in I and W" as drawn. 30 having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two differ (I) ent carbonatoms of a parent alkene. Typical alkenylene radi wherein: cals include, but are not limited to, 1.2-ethylene W is (-CH=CH-). 35 Alkynylene' refers to an unsaturated, branched or straight chain or cyclic hydrocarbon radical of 2-18 carbonatoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two differ ent carbonatoms of a parent alkyne. Typical alkynylene radi 40 cals include, but are not limited to, acetylene ( -C=C- ), propargyl (—CH2C=C ), and 4-pentynyl (-CHCHCHC=CH). The term “alkoxy” or “alkyloxy, as used herein, refers to For example, the P" group is connected to the imidazole an alkyl group attached to the parent molecular moiety group of W, and the P' group is connected to the pentacy 45 through an oxygen atom. clic ring system of W". The term “alkoxycarbonyl, as used herein, refers to an Alkyl is C-C hydrocarbon containing normal, second alkoxy group attached to the parent molecular moiety through ary, tertiary or cyclic carbon atoms. Examples are methyl a carbonyl group. (Me, —CH), ethyl (Et, —CHCH), 1-propyl (n-Pr, n-pro The term “cycloalkyl, as used herein, refers to a saturated pyl, —CH2CH2CH), 2-propyl (i-Pr. i-propyl, —CH(CH)), 50 monocyclic, hydrocarbon ring system having three to seven 1-butyl (n-Bu, n-butyl, —CH2CH2CHCH), 2-methyl-1- carbonatoms and Zero heteroatoms. Representative examples propyl (1-Bu, i-butyl, -CH-CH(CH3)2), 2-butyl (s-Bu, s-bu of cycloalkyl groups include, but are not limited to, cyclopro tyl, —CH(CH)CHCH), 2-methyl-2-propyl (t-Bu, t-butyl, pyl, cyclopentyl, and cyclohexyl. The cycloalkyl groups of —C(CH)4), 1-pentyl (n-pentyl, -CH2CH2CH2CHCH), the present disclosure are optionally substituted with one, 2-pentyl ( CH(CH)CHCHCH), 3-pentyl ( CH 55 two, three, four, or five substituents independently selected (CHCH)), 2-methyl-2-butyl ( C(CH),CHCH), 3-me from alkoxy, alkyl, aryl, cyano, halo, haloalkoxy, haloalkyl, thyl-2-butyl ( CH(CH)CH(CH)), 3-methyl-1-butyl heterocyclyl, hydroxy, hydroxyalkyl, nitro, and —NR'R'' (—CH2CHCH(CH)), 2-methyl-1-butyl ( CHCH(CH) wherein the aryl and the heterocyclyl are further optionally CHCH), 1-hexyl ( CHCH2CH2CH2CHCH), 2-hexyl substituted with one, two, or three substituents independently (-CH(CH)CHCHCHCH), 3-hexyl ( CH(CHCH.) 60 selected from alkoxy, alkyl, cyano, halo, haloalkoxy, (CHCHCH)), 2-methyl-2-pentyl(—C(CH) haloalkyl, hydroxy, and nitro. CHCHCH), 3-methyl-2-pentyl ( CH(CH)CH(CH) The term “cycloalkylcarbonyl as used herein, refers to a CHCH), 4-methyl-2-pentyl ( CH(CH)CHCH(CH)), cycloalkyl group attached to the parent molecular moiety 3-methyl-3-pentyl ( C(CH)(CHCH)), 2-methyl-3-pen through a carbonyl group. tyl ( CH(CHCH)CH(CH)), 2,3-dimethyl-2-butyl ( C 65 The term “cycloalkyloxy.” as used herein, refers to a (CH)-CH(CH)), 3.3-dimethyl-2-butyl ( CH(CH)C cycloalkyl group attached to the parent molecular moiety (CH), and cyclopropylmethyl through an oxygen atom. US 8,940,718 B2 7 8 The term “cycloalkyloxycarbonyl, as used herein, refers Examples of heterocycles include by way of example and to a cycloalkyloxy group attached to the parent molecular not limitation pyridyl, dihydropyridyl, tetrahydropyridyl (pi moiety through a carbonyl group. peridyl), thiazolyl, tetrahydrothiophenyl, sulfur oxidized tet Aryl means a monovalent aromatic hydrocarbon radical rahydrothiophenyl, pyrimidinyl, furanyl, thienyl, pyrrolyl, of6-20 carbonatoms derived by the removal of one hydrogen pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, thianaphtha atom from a single carbon atom of a parent aromatic ring lenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzimi system. Typical aryl groups include, but are not limited to, dazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, 2-pyrrolido radicals derived from benzene, substituted benzene, naphtha nyl, pyrrolinyl, tetrahydrofuranyl, tetrahydroquinolinyl, lene, anthracene, biphenyl, and the like. tetrahydroisoquinolinyl, decahydroquinolinyl, octahydroiso Arylalkyl refers to an acyclic alkyl radical in which one 10 quinolinyl, azocinyl, triazinyl, 6H-1,2,5-thiadiazinyl, of the hydrogen atoms bonded to a carbon atom, typically a 2H,6H-1.5.2-dithiazinyl, thienyl, thianthrenyl, pyranyl, terminal or sp' carbon atom, is replaced with an aryl radical. isobenzofuranyl, chromenyl, Xanthenyl, phenoxathinyl, Typical arylalkyl groups include, but are not limited to, ben 2H-pyrrolyl, isothiazolyl, isoxazolyl pyrazinyl, pyridazinyl, Zyl. 2-phenylethan-1-yl, naphthylmethyl 2-naphthylethan-1- indolizinyl, isoindolyl, 3H-indolyl, 1H-indazolyl, purinyl, yl, naphthobenzyl, 2-naphthophenylethan-1-yl and the like. 15 4H-quinolizinyl, phthalazinyl, naphthyridinyl, quinoxalinyl, The arylalkyl group comprises 6 to 20 carbonatoms, e.g., the quinazolinyl, cinnolinyl, pteridinyl, 4H-carbazolyl, carba alkyl moiety, including alkanyl, alkenyl or alkynyl groups, of Zolyl, B-carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, the arylalkyl group is 1 to 6 carbon atoms and the aryl moiety phenanthrolinyl, phenazinyl, phenothiazinyl, furazanyl, phe is 5 to 14 carbon atoms. noxazinyl, isochromanyl, chromanyl, imidazolidinyl, imida “Substituted alkyl, “substituted aryl', and “substituted Zolinyl, pyrazolidinyl, pyrazolinyl, piperazinyl, indolinyl, arylalkyl mean alkyl, aryl, and arylalkyl respectively, in isoindolinyl, quinuclidinyl, morpholinyl, oxazolidinyl, ben which one or more hydrogen atoms are each independently Zotriazolyl, benzisoxazolyl, OXindolyl, benzoxazolinyl, isati replaced with a non-hydrogen Substituent. Typical Substitu noyl, and bis-tetrahydrofuranyl: ents include, but are not limited to: halo (e.g. F, Cl, Br, I), —R, 25 OR, —SR, NR = CF, -C=Cls, —OCF, —CN, - NO, N(R)C(=O)R, C(=O)R, OC(=O)R, —C(O)CR, —C(=O)NRR, S(–O)R, S(=O),OR, —S(=O).R, OS(=O),OR, S(=O)NRR, and each R is independently —H, alkyl, aryl, arylalkyl, or heterocycle. 30 Alkylene, alkenylene, and alkynylene groups may also be similarly substituted. The term “optionally substituted in reference to a particu By way of example and not limitation, carbon bonded lar moiety of the compound of formula I, (e.g., an optionally heterocycles are bonded at position 2, 3, 4, 5, or 6 of a Substituted aryl group) refers to a moiety having 0, 1, 2, or 35 pyridine, position 3, 4, 5, or 6 of a pyridazine, position 2, 4, 5, more Substituents. or 6 of a pyrimidine, position 2, 3, 5, or 6 of a pyrazine, The symbol "----- in a ring structure means that a bond is position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, a single or double bond. In a non-limiting example, thiophene, pyrrole or tetrahydropyrrole, position 2, 4, or 5 of an oxazole, imidazole or thiazole, position 3, 4, or 5 of an 40 isoxazole, pyrazole, or isothiazole, position 2 or 3 of an L L aziridine, position 2, 3, or 4 of anaZetidine, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline or position 1, 3, 4, 5, 6, 7, or 8 of an can be { O isoquinoline. Still more typically, carbon bonded hetero b 2. \, 2. cycles include 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, L 45 6-pyridyl, 3-pyridazinyl, 4-pyridaZinyl, 5-pyridazinyl, 6-py ridazinyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, 6-py rimidinyl, 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl. By way of example and not limitation, nitrogen bonded "Haloalkyl as used herein includes an alkyl group substi 50 heterocycles are bonded at position 1 of an aziridine, aZeti tuted with one or more halogens (e.g. F, Cl, Br, or I). Repre dine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imida sentative examples of haloalkyl include trifluoromethyl, 2.2, Zole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, 2-trifluoroethyl, and 2.2.2-trifluoro-1-(trifluoromethyl)ethyl. pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, pipera “Heterocycle' or "heterocyclyl as used herein includes by Zine, indole, indoline, 1H-indazole, position 2 of a isoindole, way of example and not limitation these heterocycles 55 or isoindoline, position 4 of a morpholine, and position 9 of a described in Paquette, Leo A.; Principles of Modern Hetero carbazole, or 3-carboline. Still more typically, nitrogen cyclic Chemistry (W.A. Benjamin, New York, 1968), particu bonded heterocycles include 1-aziridyl, 1-azetedyl, 1-pyrro larly Chapters 1, 3, 4, 6, 7, and 9: The Chemistry of Hetero lyl, 1-imidazolyl, 1-pyrazolyl, and 1-piperidinyl. cyclic Compounds, A Series of Monographs” (John Wiley & “Carbocycle” refers to a saturated, unsaturated or aromatic Sons, New York, 1950 to present), in particular Volumes 13, 60 ring having up to about 25 carbon atoms. Typically, a car 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82:5566. In bocycle has about 3 to 7 carbon atoms as a monocycle, about one specific embodiment, “heterocycle' includes a “car 7 to 12 carbon atoms as a bicycle, and up to about 25 carbon bocycle' as defined herein, wherein one or more (e.g. 1, 2, 3, atoms as a polycycle. Monocyclic carbocycles typically have or 4) carbonatoms have been replaced with a heteroatom (e.g. 3 to 6 ring atoms, still more typically 5 or 6 ring atoms. O, N, or S). The term heterocycle also includes "heteroaryl 65 Bicyclic carbocycles typically have 7 to 12 ring atoms, e.g., which is a heterocycle wherein at least one heterocyclic rings arranged as a bicyclo4.5, 5.5, 5.6 or 6.6 system, or 9 or is aromatic. 10 ring atoms arranged as a bicyclo5.6 or 6.6 system. The US 8,940,718 B2 10 term carbocycle includes “cycloalkyl” which is a saturated or “Design and Application of ' in A Textbook of Drug unsaturated carbocycle. Examples of monocyclic car Design and Development (1991), P. Krogsgaard-Larsen and bocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1-cy H. Bundgaard, Eds. Harwood Academic Publishers, pp. 113 clopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, 191). Enzymes which are capable of an enzymatic activation cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclo 5 mechanism with the prodrug compounds of the disclosure hex-3-enyl, phenyl, spiryland naphthyl. include, but are not limited to, amidases, esterases, microbial The term "amino.” as used herein, refers to NH. enzymes, phospholipases, cholinesterases, and phosphases. The term "chiral refers to molecules which have the prop Prodrug moieties can serve to enhance solubility, absorption erty of non-Superimposability of the mirror image partner, and lipophilicity to optimize drug delivery, bioavailability while the term “achiral” refers to molecules which are super 10 and efficacy. A prodrug moiety may include an active metabo imposable on their mirror image partner. lite or drug itself. The term “stereoisomers' refers to compounds which have Exemplary prodrug moieties include the hydrolytically identical chemical constitution, but differ with regard to the sensitive or labile acyloxymethyl esters —CHOC(=O)R’ arrangement of the atoms or groups in space. and acyloxymethyl carbonates —CHC(=O)CR where “Diastereomer' refers to a stereoisomer with two or more 15 R’ is C-C alkyl, C-C substituted alkyl, Co-Co aryl or centers of chirality and whose molecules are not mirror Co-Co Substituted aryl. The acyloxyalkyl ester was first used images of one another. Diastereomers have different physical as a prodrug strategy for carboxylic acids and then applied to properties, e.g., melting points, boiling points, spectral prop phosphates and phosphonates by Farquhar et al. (1983) J. erties, and reactivities. Mixtures of diastereomers may sepa Pharm. Sci. 72: 324; also U.S. Pat. Nos. 4,816,570, 4,968, rate under high resolution analytical procedures such as elec 788, 5,663,159 and 5,792,756. Subsequently, the acyloxy trophoresis and chromatography. alkyl ester was used to deliver phosphonic acids across cell "Enantiomers' refer to two stereoisomers of a compound membranes and to enhance oral bioavailability. A close vari which are non-Superimposable mirror images of one another. ant of the acyloxyalkyl ester, the alkoxycarbonyloxyalkyl The term “treatment” or “treating to the extent it relates to ester (carbonate), may also enhance oral bioavailability as a a disease or condition includes preventing the disease or 25 prodrug moiety in the compounds of the combinations of the condition from occurring, inhibiting the disease or condition, disclosure. An exemplary acyloxymethyl ester is pivaloy eliminating the disease or condition, and/or relieving one or loxymethoxy, (POM) —CHC(=O)C(CH). An exemplary more symptoms of the disease or condition. acyloxymethyl carbonate prodrug moiety is pivaloyloxym Stereochemical definitions and conventions used herein ethylcarbonate (POC)—CHC(=O)CC(CH). generally follow S.P. Parker, Ed., McGraw-Hill Dictionary of 30 Aryl esters of phosphorus groups, especially phenyl esters, Chemical Terms (1984) McGraw-Hill Book Company, New are reported to enhance oral bioavailability (De Lombaert et York; and Eliel, E. and Wilen, S., Stereochemistry of Organic al. (1994).J. Med Chem. 37: 498). Phenyl esters containing a Compounds (1994) John Wiley & Sons, Inc., New York. carboxylic ester ortho to a phosphate have also been Many organic compounds exist in optically active forms, i.e., described (Khamnei and Torrence, (1996) J. Med. Chem. they have the ability to rotate the plane of plane-polarized 35 39:4109-4115). Benzyl esters are reported to generate parent light. In describing an optically active compound, the prefixes phosphonic acids. In some cases, Substituents at the ortho- or (D and L) or (R and S) are used to denote the absolute para-position may accelerate the hydrolysis. Benzyl analogs configuration of the molecule about its chiral center(s). The with an acylated phenol or an alkylated phenol may generate prefixes d and 1 or (+) and (-) are employed to designate the the phenolic compound through the action of enzymes, e.g., sign of rotation of plane-polarized light by the compound, 40 esterases, oxidases, etc., which in turn undergoes cleavage at with (-) or 1 meaning that the compound is levorotatory. A the benzylic C-O bond to generate phosphoric acid and a compound prefixed with (+) ord is dextrorotatory. For a given quinone methide intermediate. Examples of this class of pro chemical structure, these stereoisomers are identical except drugs are described by Mitchell et al. (1992) J. Chem. Soc. that they are mirror images of one another. A specific Stere Perkin Trans. II 2345; Glazier WO91/19721. Still other ben oisomer may also be referred to as an enantiomer, and a 45 Zylic prodrugs have been described containing a carboxylic mixture of Such isomers is often called an enantiomeric mix ester-containing group attached to the benzylic methylene ture. A 50:50 mixture of enantiomers is referred to as a race (Glazier WO 91/19721). Thio-containing prodrugs are mic mixture or a racemate, which may occur where there has reported to be useful for the intracellular delivery of phos been no stereoselection or Stereospecificity in a chemical phonate drugs. These proesters contain an ethylthio group in reaction or process. The terms “racemic mixture' and “race 50 which the thiol group is eitheresterified with an acyl group or mate” refer to an equimolar mixture of two enantiomeric combined with another thiol group to form a disulfide. Dees species, devoid of optical activity. The disclosure includes all terification or reduction of the disulfide generates the free thio Stereoisomers of the compounds described herein. intermediate which subsequently breaks down to the phos phoric acid and episulfide (Puechet al. (1993) Antiviral Res., Prodrugs 55 22: 155-174; Benzaria et al. (1996).J. Med. Chem. 39: 4958). The term “prodrug” as used herein refers to any compound Protecting Groups that when administered to a biological system generates a compound of the disclosure that inhibits HCV activity (“the In the context of the present disclosure, protecting groups active inhibitory compound'). The compound may beformed 60 include prodrug moieties and chemical protecting groups. from the prodrug as a result of: (i) spontaneous chemical “Protecting group' refers to a moiety of a compound that reaction(s), (ii) enzyme catalyzed chemical reaction(s), (iii) masks or alters the properties of a functional group or the photolysis, and/or (iv) metabolic chemical reaction(s). properties of the compound as a whole. Chemical protecting “Prodrug moiety” refers to a labile functional group which groups and strategies for protection/deprotection are well separates from the active inhibitory compound during 65 known in the art. See e.g., Protective Groups in Organic metabolism, systemically, inside a cell, by hydrolysis, enzy Chemistry. Theodora W. Greene, John Wiley & Sons, Inc., matic cleavage, or by some other process (Bundgaard, Hans, New York, 1991. Protecting groups are often utilized to mask US 8,940,718 B2 11 12 the reactivity of certain functional groups, to assist in the boxylic acid, phosphonic acid, phosphonate, Sulfonic acid efficiency of desired chemical reactions, e.g., making and and other protecting groups for acids see Greene as set forth breaking chemical bonds in an ordered and planned fashion. below. Protection of functional groups of a compound alters other By way of example and not limitation, variables described physical properties besides the reactivity of the protected 5 herein may recursive Substituents in certain embodiments. functional group, Such as the polarity, lipophilicity (hydro Typically, each of these may independently occur 20, 19, 18, phobicity), and other properties which can be measured by 17, 16, 15, 14, 13, 12, 11, 10,9,8,7,6, 5, 4, 3, 2, 1, or 0, times common analytical tools. Chemically protected intermedi in a given embodiment. More typically, each of these may independently occur 12 or fewer times in a given embodi ates may themselves be biologically active or inactive. ment. Whenever a compound described herein is substituted Protected compounds may also exhibit altered, and in some 10 with more than groups may be the same or different, i.e., each cases, optimized properties in vitro and in vivo, Such as pas group is independently selected. Wavy lines indicate the site sage through cellular membranes and resistance to enzymatic of covalent bond attachments to the adjoining groups, moi degradation or sequestration. In this role, protected com eties, or atoms. pounds with intended therapeutic effects may be referred to as In one embodiment of the disclosure, the compound is in an prodrugs. Another function of a protecting group is to convert 15 isolated and purified form. Generally, the term "isolated and the parental drug into a prodrug, whereby the parental drug is purified” means that the compound is substantially free from released upon conversion of the prodrug in vivo. Because biological materials (e.g. blood, tissue, cells, etc.). In one active prodrugs may be absorbed more effectively than the specific embodiment of the disclosure, the term means that parental drug, prodrugs may possess greater potency in vivo the compound or conjugate of the disclosure is at least about than the parental drug. Protecting groups are removed either 50 wt.% free from biological materials; in another specific in vitro, in the instance of chemical intermediates, or in vivo, embodiment, the term means that the compound or conjugate in the case of prodrugs. With chemical intermediates, it is not of the disclosure is at least about 75 wt.% free from biological particularly important that the resulting products after depro materials; in another specific embodiment, the term means tection, e.g., alcohols, be physiologically acceptable, that the compound or conjugate of the disclosure is at least although in general it is more desirable if the products are 25 about 90 wt.% free from biological materials; in another pharmacologically innocuous. specific embodiment, the term means that the compound or Protecting groups are available, commonly known and conjugate of the disclosure is at least about 98 wt.% free from biological materials; and in another embodiment, the term used, and are optionally used to prevent side reactions with means that the compound or conjugate of the disclosure is at the protected group during synthetic procedures, i.e. routes or least about 99 wt.% free from biological materials. In another methods to prepare the compounds of the disclosure. For the 30 specific embodiment, the disclosure provides a compound or most part the decision as to which groups to protect, when to conjugate of the disclosure that has been synthetically pre do so, and the nature of the chemical protecting group “PG” pared (e.g., ex vivo). will be dependent upon the chemistry of the reaction to be protected against (e.g., acidic, basic, oxidative, reductive or Stereoisomers other conditions) and the intended direction of the synthesis. 35 PGs do not need to be, and generally are not, the same if the The compounds of the disclosure may have chiral centers, compound is substituted with multiple PGs. In general, PG e.g., chiral carbon or phosphorus atoms. The compounds of will be used to protect functional groups such as carboxyl, the disclosure thus include racemic mixtures of all stereoiso hydroxyl, thio, or amino groups and to thus prevent side mers, including enantiomers, diastereomers, and atropiso reactions or to otherwise facilitate the synthetic efficiency. 40 mers. In addition, the compounds of the disclosure include The order of deprotection to yield free deprotected groups is enriched or resolved optical isomers at any or all asymmetric, dependent upon the intended direction of the synthesis and chiral atoms. In other words, the chiral centers apparent from the reaction conditions to be encountered, and may occur in the depictions are provided as the chiral isomers or racemic any order as determined by the artisan. mixtures. Both racemic and diastereomeric mixtures, as well Various functional groups of the compounds of the disclo 45 as the individual optical isomers isolated or synthesized, Sub Sure may be protected. For example, protecting groups for stantially free of their enantiomeric or diastereomeric part —OH groups (whether hydroxyl, carboxylic acid, phospho ners, are all within the scope of the disclosure. The racemic nic acid, or other functions) include “ether- or ester-forming mixtures are separated into their individual, Substantially groups”. Ether- or ester-forming groups are capable of func optically pure isomers through well-known techniques such tioning as chemical protecting groups in the synthetic 50 as, for example, the separation of diastereomeric salts formed schemes set forth herein. However, some hydroxyl and thio with optically active adjuncts, e.g., acids or bases followed by protecting groups are neither ether-norester-forming groups, conversion back to the optically active Substances. In most as will be understood by those skilled in the art, and are instances, the desired optical isomer is synthesized by means included with amides, discussed below. of stereospecific reactions, beginning with the appropriate A very large number of hydroxyl protecting groups and 55 Stereoisomer of the desired starting material. amide-forming groups and corresponding chemical cleavage The compounds of the disclosure can also exist as tauto reactions are described in Protective Groups in Organic Syn meric isomers in certain cases. Although only one tautomer thesis, Theodora W. Greene (John Wiley & Sons, Inc., New may be depicted, all such forms are contemplated within the York, 1991, ISBN 0-471-62301-6) (“Greene'). See also Scope of the disclosure. For example, ene-amine tautomers Kocienski, Philip J.; Protecting Groups (Georg Thieme Ver 60 can exist for purine, pyrimidine, imidazole, guanidine, ami lag Stuttgart, New York, 1994), which is incorporated by dine, and tetrazole systems and all their possible tautomeric reference in its entirety herein. In particular Chapter 1, Pro forms are within the scope of the disclosure. tecting Groups: An Overview, pages 1-20, Chapter 2, Hydroxyl Protecting Groups, pages 21-94, Chapter 3, Diol Salts and Hydrates Protecting Groups, pages 95-117, Chapter 4, Carboxyl Pro 65 tecting Groups, pages 118-154, Chapter 5, Carbonyl Protect Examples of physiologically or pharmaceutically accept ing Groups, pages 155-184. For protecting groups for car able salts of the compounds of the disclosure include salts US 8,940,718 B2 13 14 derived from an appropriate base. Such as an alkali metal (for field and include stable free radicals, fluorophores, radioiso example, Sodium), an alkaline earth metal (for example, mag topes, enzymes, chemiluminescent groups and chromogens. nesium), ammonium and NX (wherein X is C-C alkyl). The compounds herein are labeled in conventional fashion Physiologically acceptable salts of a hydrogen atom or an using functional groups such as hydroxyl or amino. In one amino group include salts of organic carboxylic acids such as 5 embodiment the disclosure provides a compound of any one acetic, benzoic, lactic, fumaric, tartaric, maleic, malonic, of formulae (I) and (A1)-(A4) that comprises or that is bound malic, isethionic, lactobionic and Succinic acids; organic Sul or linked to one or more detectable labels. Within the context fonic acids, such as methanesulfonic, ethanesulfonic, benze of the disclosure samples suspected of containing HCV nesulfonic and p-toluenesulfonic acids; and inorganic acids, include natural or man-made materials such as living organ Such as hydrochloric, Sulfuric, phosphoric and Sulfamic acids. 10 isms; tissue or cell cultures; biological samples such as bio Physiologically acceptable salts of a compound of a hydroxy logical material samples (blood, serum, urine, cerebrospinal group include the anion of said compound in combination with a suitable cation such as Na' and NX (wherein X is fluid, tears, sputum, saliva, tissue samples, and the like); independently selected from H or a C-C alkyl group). laboratory samples; food, water, or air samples; bioproduct For therapeutic use, salts of active ingredients of the com 15 samples Such as extracts of cells, particularly recombinant pounds of the disclosure will typically be physiologically cells synthesizing a desired glycoprotein; and the like. Typi acceptable, i.e. they will be salts derived from a physiologi cally the sample will be suspected of containing HCV. cally acceptable acid or base. However, salts of acids or bases Samples can be contained in any medium including water and which are not physiologically acceptable may also find use, organic solvent/water mixtures. Samples include living for example, in the preparation or purification of a physiologi organisms such as humans, and man made materials such as cally acceptable compound. All salts, whether or not derived cell cultures. from a physiologically acceptable acid or base, are within the The treating step of the disclosure comprises adding the Scope of the present disclosure. compound of the disclosure to the sample or it comprises Metal salts typically are prepared by reacting the metal adding a precursor of the composition to the sample. The hydroxide with a compound of this disclosure. Examples of 25 addition step comprises any method of administration as metal salts which are prepared in this way are salts containing described above. Li, Na', and K". A less soluble metal salt can be precipitated If desired, the activity of HCV after application of the from the solution of a more soluble salt by addition of the compound can be observed by any method including direct Suitable metal compound. and indirect methods of detecting HCV activity. Quantitative, In addition, salts may be formed from acid addition of 30 qualitative, and semiquantitative methods of determining certain organic and inorganic acids, e.g., HCl, HBr, HSO HCV activity are all contemplated. Typically one of the HPO or organic sulfonic acids, to basic centers, typically screening methods described above are applied, however, any amines, or to acidic groups. Finally, it is to be understood that other method such as observation of the physiological prop the compositions herein comprise compounds of the disclo erties of a living organism are also applicable. Sure in their un-ionized, as well as Zwitterionic form, and 35 Many organisms contain HCV. The compounds of this combinations with Stoichiometric amounts of water as in disclosure are useful in the treatment or prophylaxis of con hydrates. ditions associated with HCV activation in animals or in man. Also included within the scope of this disclosure are the However, in screening compounds capable of inhibiting salts of the parental compounds with one or more amino HCV activity it should be kept in mind that the results of acids. Any of the natural or unnatural amino acids are Suit 40 enzyme assays may not always correlate with cell culture able, especially the naturally-occurring amino acids found as assays. Thus, a cell based assay should typically be the pri protein components, although the amino acid typically is one mary screening tool. bearing a side chain with a basic or acidic group, e.g., lysine, arginine or glutamic acid, or a neutral group Such as glycine, Pharmaceutical Formulations serine, threonine, alanine, isoleucine, or leucine. 45 The compounds of this disclosure are formulated with Methods of Inhibition of HCV conventional carriers and excipients, which will be selected in accord with ordinary practice. Tablets will contain excipients, Another aspect of the disclosure relates to methods of glidants, fillers, binders and the like. Aqueous formulations inhibiting the activity of HCV comprising the step of treating 50 are prepared in sterile form, and when intended for delivery a sample Suspected of containing HCV with a compound or by other than oral administration generally will be isotonic. composition of the disclosure. All formulations will optionally contain excipients such as Compounds of the disclosure may act as inhibitors of HCV. those set forth in the Handbook of Pharmaceutical Excipients as intermediates for such inhibitors or have other utilities as (1986). Excipients include ascorbic acid and other antioxi described below. The inhibitors will generally bind to loca 55 dants, chelating agents such as EDTA, carbohydrates Such as tions on the Surface or in a cavity of the liver. Compounds dextrin, hydroxyalkylcellulose, hydroxyalkylmethylcellu binding in the liver may bind with varying degrees of revers lose, stearic acid and the like. The pH of the formulations ibility. Those compounds binding substantially irreversibly ranges from about 3 to about 11, but is ordinarily about 7 to are ideal candidates for use in this method of the disclosure. 10. Typically, the compound will be administered in a dose Once labeled, the substantially irreversibly binding com 60 from 0.01 milligrams to 2 grams. In one embodiment, the pounds are useful as probes for the detection of HCV. Accord dose will be from about 10 milligrams to 450 milligrams. In ingly, the disclosure relates to methods of detecting NS3 in a another embodiment, the dosage will be from about 25 to sample Suspected of containing HCV comprising the steps of about 250 milligrams. In another embodiment, the dosage treating a sample Suspected of containing HCV with a com will be about 50 or 100 milligrams. In one embodiment, the position comprising a compound of the disclosure bound to a 65 dosage will be about 100 milligrams. It is contemplated that label; and observing the effect of the sample on the activity of the compound may be administered once, twice or three times the label. Suitable labels are well known in the diagnostics a day. US 8,940,718 B2 15 16 While it is possible for the active ingredients to be admin and an oil. Preferably, a hydrophilic emulsifier is included istered alone it may be preferable to present them as pharma together with a lipophilic emulsifier which acts as a stabilizer. ceutical formulations. The formulations, both for veterinary It is also preferred to include both an oil and a fat. Together, and for human use, of the disclosure comprise at least one the emulsifier(s) with or without stabilizer(s) make up the active ingredient, as above defined, together with one or more so-called emulsifying wax, and the wax together with the oil acceptable carriers therefor and optionally other therapeutic and fat make up the so-called emulsifying ointment base ingredients. The carrier(s) must be “acceptable' in the sense which forms the oily dispersed phase of the cream formula of being compatible with the other ingredients of the formu tions. lation and physiologically innocuous to the recipient thereof. Emulgents and emulsion stabilizers suitable for use in the The formulations include those suitable for the foregoing 10 formulation of the disclosure include Tween(R) 60, SpanR 80. administration routes. The formulations may conveniently be cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyc presented in unit dosage form and may be prepared by any of eryl mono-stearate and sodium lauryl Sulfate. the methods well known in the art of pharmacy. Techniques The choice of suitable oils or fats for the formulation is and formulations generally are found in Remington's Phar based on achieving the desired cosmetic properties. The maceutical Sciences (Mack Publishing Co., Easton, Pa.). 15 cream should preferably be a non-greasy, non-staining and Such methods include the step of bringing into association the washable product with Suitable consistency to avoid leakage active ingredient with the carrier which constitutes one or from tubes or other containers. more accessory ingredients. In general the formulations are Straight or branched chain, mono- or dibasic alkyl esters prepared by uniformly and intimately bringing into associa Such as di-isoadipate, isocetyl Stearate, propylene glycol tion the active ingredient with liquid carriers or finely divided diester of coconut fatty acids, isopropyl myristate, decyl ole Solid carriers or both, and then, if necessary, shaping the ate, isopropyl palmitate, butyl Stearate, 2-ethylhexyl palmi product. tate or a blend of branched chain esters known as Crodamol Formulations of the present disclosure suitable for oral CAP may be used, the last three being preferred esters. These administration may be presented as discrete units such as may be used alone or in combination depending on the prop capsules, cachets or tablets each containing a predetermined 25 erties required. Alternatively, high melting point lipids Such amount of the active ingredient; as a powder or granules; as a as white soft paraffin and/or liquid paraffin or other mineral Solution or a Suspension in an aqueous or non-aqueous liquid; oils are used. or as an oil-in-water liquid emulsion or a water-in-oil liquid Pharmaceutical formulations according to the present dis emulsion. The active ingredient may also be administered as closure comprise one or more compounds of the disclosure a bolus, electuary or paste. 30 together with one or more pharmaceutically acceptable car A tablet is made by compression or molding, optionally riers or excipients and optionally other therapeutic agents. with one or more accessory ingredients. Compressed tablets Pharmaceutical formulations containing the active ingredient may be prepared by compressing in a Suitable machine the may be in any form suitable for the intended method of active ingredient in a free-flowing form Such as a powder or administration. When used for oral use for example, tablets, granules, optionally mixed with a binder, lubricant, inert dilu 35 troches, lozenges, aqueous or oil Suspensions, dispersible ent, preservative, Surface active or dispersing agent. Molded powders or granules, emulsions, hard or soft capsules, syrups tablets may be made by molding in a suitable machine a or elixirs may be prepared. Compositions intended for oral mixture of the powdered active ingredient moistened with an use may be prepared according to any method known to the inert liquid diluent. The tablets may optionally be coated or art for the manufacture of pharmaceutical compositions and scored and optionally are formulated so as to provide slow or 40 Such compositions may contain one or more agents including controlled release of the active ingredient therefrom. Sweetening agents, flavoring agents, coloring agents and pre For administration to the eye or other external tissues e.g., serving agents, in order to provide a palatable preparation. mouth and skin, the formulations are preferably applied as a Tablets containing the active ingredient in admixture with topical ointment or cream containing the active ingredient(s) non-toxic pharmaceutically acceptable excipient which are in an amount of, for example, 0.075 to 20% w/w (including 45 suitable for manufacture of tablets are acceptable. These active ingredient(s) in a range between 0.1% and 20% in excipients may be, for example, inert diluents, such as cal increments of 0.1% w/w such as 0.6% w/w, 0.7% w/w, etc.), cium or sodium carbonate, lactose, lactose monohydrate, preferably 0.2 to 15% w/w and most preferably 0.5 to 10% croScarmellose Sodium, povidone, calcium or sodium phos w/w. When formulated in an ointment, the active ingredients phate; granulating and disintegrating agents, such as maize may be employed with either a paraffinic or a water-miscible 50 starch, or alginic acid; binding agents, such as cellulose, ointment base. Alternatively, the active ingredients may be microcrystalline cellulose, starch, gelatin oracacia; and lubri formulated in a cream with an oil-in-water cream base. cating agents. Such as magnesium Stearate, Stearic acid or talc. If desired, the aqueous phase of the cream base may Tablets may be uncoated or may be coated by known tech include, for example, at least 30% w/w of a polyhydric alco niques including microencapsulation to delay disintegration hol, i.e. an alcohol having two or more hydroxyl groups such 55 and adsorption in the gastrointestinal tract and thereby pro as propylene glycol, butane 1,3-diol, mannitol, Sorbitol, glyc vide a Sustained action over a longer period. For example, a erol and polyethylene glycol (including PEG 400) and mix time delay material Such as glyceryl monostearate or glyceryl tures thereof. The topical formulations may desirably include distearate alone or with a wax may be employed. a compound which enhances absorption or penetration of the Formulations for oral use may be also presented as hard active ingredient through the skin or other affected areas. 60 gelatin capsules where the active ingredient is mixed with an Examples of such dermal penetration enhancers include dim inert Solid diluent, for example calcium phosphate or kaolin, ethyl Sulphoxide and related analogs. or as Soft gelatin capsules wherein the active ingredient is The oily phase of the emulsions of this disclosure may be mixed with water oran oil medium, Such as peanut oil, liquid constituted from known ingredients in a known manner. paraffin or olive oil. While the phase may comprise merely an emulsifier (other 65 Aqueous Suspensions of the disclosure contain the active wise known as an emulgent), it desirably comprises a mixture materials in admixture with excipients Suitable for the manu of at least one emulsifier with a fat or an oil or with both a fat facture of aqueous Suspensions. Such excipients include a US 8,940,718 B2 17 18 Suspending agent, such as Sodium carboxymethylcellulose, of administration. For example, a time-release formulation methylcellulose, hydroxypropyl methylcellulose, sodium intended for oral administration to humans may contain alginate, polyvinylpyrrolidone, gum tragacanthandgum aca approximately 1 to 1000 mg of active material compounded cia, and dispersing or wetting agents such as a naturally with an appropriate and convenient amount of carrier material occurring phosphatide (e.g., lecithin), a condensation product 5 which may vary from about 5 to about 95% of the total of an alkylene oxide with a fatty acid (e.g., polyoxyethylene compositions (weight: weight). The pharmaceutical compo Stearate), a condensation product of ethylene oxide with a sition can be prepared to provide easily measurable amounts long chain aliphatic alcohol (e.g., heptadecaethyleneoxycet for administration. For example, an aqueous solution anol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., 10 intended for intravenous infusion may contain from about 3 to polyoxyethylene Sorbitan monooleate). The aqueous Suspen 500 ug of the active ingredient per milliliter of solution in sion may also contain one or more preservatives such as ethyl order that infusion of a suitable volume at a rate of about 30 or n-propyl p-hydroxy-benzoate, one or more coloring mL/hr can occur. agents, one or more flavoring agents and one or more Sweet Formulations suitable for administration to the eye include ening agents. Such as Sucrose or saccharin. 15 eye drops wherein the active ingredient is dissolved or Sus Oil Suspensions may be formulated by Suspending the pended in a suitable carrier, especially an aqueous solvent for active ingredient in a vegetable oil. Such as arachis oil, olive the active ingredient. The active ingredient is preferably oil, Sesame oil or coconut oil, or in a mineral oil such as liquid present in such formulations in a concentration of 0.5 to 20%, paraffin. The oral Suspensions may contain a thickening advantageously 0.5 to 10% particularly about 1.5% w/w. agent, such as beeswax, hard paraffin or cetyl alcohol. Sweet- 20 Formulations suitable for topical administration in the ening agents, such as those set forth above, and flavoring mouth include lozenges comprising the active ingredient in a agents may be added to provide a palatable oral preparation. flavored basis, usually Sucrose and acacia or tragacanth; pas These compositions may be preserved by the addition of an tilles comprising the active ingredient in an inert basis such as antioxidant Such as ascorbic acid. gelatin and glycerin, or Sucrose and acacia; and mouthwashes Dispersible powders and granules of the disclosure suit- 25 comprising the active ingredient in a suitable liquid carrier. able for preparation of an aqueous Suspension by the addition Formulations for rectal administration may be presented as of water provide the active ingredient in admixture with a a Suppository with a Suitable base comprising for example dispersing or wetting agent, a Suspending agent, and one or cocoa butter or a salicylate. more preservatives. Suitable dispersing or wetting agents and Formulations Suitable for intrapulmonary or nasal admin Suspending agents are exemplified by those disclosed above. 30 istration have a particle size for example in the range of 0.1 to Additional excipients, for example Sweetening, flavoring and 500 microns (including particle sizes in a range between 0.1 coloring agents, may also be present. and 500 microns in increments such as 0.5 microns, 1 micron, The pharmaceutical compositions of the disclosure may 30 microns, 35 microns, etc.), which is administered by rapid also be in the form of oil-in-water emulsions. The oily phase inhalation through the nasal passage or by inhalation through may be a vegetable oil. Such as olive oil or arachis oil, a 35 the mouth so as to reach the alveolar sacs. Suitable formula mineral oil. Such as liquid paraffin, or a mixture of these. tions include aqueous or oily solutions of the active ingredi Suitable emulsifying agents include naturally-occurring ent. Formulations suitable for aerosol or dry powder admin gums, such as gum acacia and gum tragacanth, naturally istration may be prepared according to conventional methods occurring phosphatides, such as soybean lecithin, esters or and may be delivered with other therapeutic agents such as partial esters derived from fatty acids and hexitol anhydrides, 40 compounds heretofore used in the treatment or prophylaxis of Such as Sorbitan monooleate, and condensation products of conditions associated with HCV activity. these partial esters with ethylene oxide. Such as polyoxyeth Formulations suitable for vaginal administration may be ylene Sorbitan monooleate. The emulsion may also contain presented as pessaries, tampons, creams, gels, pastes, foams Sweetening and flavoring agents. Syrups and elixirs may be or spray formulations containing in addition to the active formulated with Sweetening agents, such as glycerol, Sorbitol 45 ingredient Such carriers as are known in the art to be appro or Sucrose. Such formulations may also contain a demulcent, priate. a preservative, a flavoring or a coloring agent. Formulations suitable for parenteral administration The pharmaceutical compositions of the disclosure may be include aqueous and non-aqueous sterile injection solutions in the form of a sterile injectable preparation, such as a sterile which may contain anti-oxidants, buffers, bacteriostats and injectable aqueous or oleaginous Suspension. This suspen- 50 solutes which render the formulation isotonic with the blood sion may be formulated according to the known art using of the intended recipient; and aqueous and non-aqueous ster those Suitable dispersing or wetting agents and Suspending ille Suspensions which may include Suspending agents and agents which have been mentioned above. The sterile inject thickening agents. able preparation may also be a sterile injectable solution or The formulations are presented in unit-dose or multi-dose Suspension in a non-toxic parenterally acceptable diluent or 55 containers, for example sealed ampoules and vials, and may Solvent. Such as a solution in 1,3-butane-diol or prepared as a be stored in a freeze-dried (lyophilized) condition requiring lyophilized powder. Among the acceptable vehicles and Sol only the addition of the sterile liquid carrier, for example vents that may be employed are water, Ringer's solution and water for injection, immediately prior to use. Extemporane isotonic sodium chloride solution. In addition, sterile fixed ous injection Solutions and Suspensions are prepared from oils may conventionally be employed as a solventor Suspend- 60 sterile powders, granules and tablets of the kind previously ing medium. For this purpose any bland fixed oil may be described. Preferred unit dosage formulations are those con employed including synthetic mono- or diglycerides. In addi taining a daily dose or unit daily Sub-dose, as herein above tion, fatty acids Such as oleic acid may likewise be used in the recited, or an appropriate fraction thereof, of the active ingre preparation of injectables. dient. The amount of active ingredient that may be combined 65 It should be understood that in addition to the ingredients with the carrier material to produce a single dosage form will particularly mentioned above the formulations of this disclo vary depending upon the host treated and the particular mode Sure may include other agents conventional in the art having US 8,940,718 B2 19 20 regard to the type of formulation in question, for example BI-1335, BI-1230, MK-7009, VBY-376, VX-500, GS-9256, those Suitable for oral administration may include flavoring GS-9451, BMS-605339, PHX-1766, AS-101, YH-5258, agents. YH5530, YH5531, ABT-450, ACH-1625, ITMN-191, The disclosure further provides veterinary compositions MK5172, MK6325, and MK2748; comprising at least one active ingredient as above defined 5 4) alpha-glucosidase 1 inhibitors, e.g., celgosivir (MX together with a veterinary carrier therefor. 3253), Miglitol, and UT-231B: Veterinary carriers are materials useful for the purpose of 5) hepatoprotectants, e.g., emericasan (IDN-6556), administering the composition and may be solid, liquid or ME-3738, GS-9450 (LB-84451), silibilin, and MitoO: gaseous materials which are otherwise inert or acceptable in 6) nucleoside or nucleotide inhibitors of HCV NS5B poly the veterinary art and are compatible with the active ingredi 10 merase, e.g., R1626, R7128 (R4048), IDX184, IDX-102, ent. These veterinary compositions may be administered BCX-4678, (NM-283), MK-0608, sofosbuvir orally, parenterally or by any other desired route. (GS-7977 (formerly PSI-7977)), and INX-189 (now Compounds of the disclosure can also be formulated to BMS986094); provide controlled release of the active ingredient to allow 7) non-nucleoside inhibitors of HCV NS5B polymerase, less frequent dosing or to improve the pharmacokinetic or 15 e.g., PF-868554, VCH-759, VCH-916, JTK-652, MK-3281, toxicity profile of the active ingredient. Accordingly, the dis GS-9190, VBY-708, VCH-222, A848837, ANA-598, closure also provides compositions comprising one or more GL60667, GL59728, A-63890, A-48773, A-48547, compounds of the disclosure formulated for Sustained or con BC-2329, VCH-796 (nesbuvir), GSK625433, BILN-1941, trolled release. XTL-2125, ABT-072, ABT-333, GS-9669, PSI-7792, and Effective dose of active ingredient depends at least on the GS-9190; nature of the condition being treated, toxicity, whether the 8) HCV NS5A inhibitors, e.g., AZD-2836 (A-831), BMS compound is being used prophylactically (lower doses), the 790052, ACH-3102, ACH-2928, MK8325, MK4882, method of delivery, and the pharmaceutical formulation, and MK8742, PSI-461, IDX719, and A-689: will be determined by the clinician using conventional dose 9) TLR-7 agonists, e.g., , 852A, GS-9524, escalation studies. 25 ANA-773, ANA-975, AZD-8848 (DSP-3025), and SM-360320; 10) cyclophillin inhibitors, e.g., DEBIO-025, Routes of Administration SCY-635, and NIM811; 11) HCV IRES inhibitors, e.g., MCI-067: One or more compounds of the disclosure (herein referred 12) pharmacokinetic enhancers, e.g., BAS-100, SPI-452, to as the active ingredients) are administered by any route 30 PF-4194477, TMC-41629, GS-9350, GS-9585, and roxyth appropriate to the condition to be treated. Suitable routes romycin; and include oral, rectal, nasal, topical (including buccal and sub 13) other drugs for treating HCV, e.g., thymosin alpha 1 lingual), vaginal and parenteral (including Subcutaneous, (Zadaxin), nitazoxanide (Alinea, NTZ), BIVN-401 (vi intramuscular, intravenous, intradermal, intrathecal and epi rostat), PYN-17 (altirex), KPE02003002, actilon (CPG dural), and the like. It will be appreciated that the preferred 35 10101), GS-9525, KRN-7000, civacir, GI-5005, XTL-6865, route may vary with for example the condition of the recipi BIT225, PTX-111, ITX2865, TT-033i, ANA971, NOV-205, ent. An advantage of the compounds of this disclosure is that tarvacin, EHC-18, VGX-410C, EMZ-702, AVI 4065, BMS they are orally bioavailable and can be dosed orally. 650032, BMS-791325, Bavituximab, MDX-1 106 (ONO HCV Combination Therapy 4538), Oglufanide, and VX-497 (merimepodlib). In another embodiment, non-limiting examples of Suitable 40 More specifically, one or more compounds as described combinations include combinations of one or more com herein may be combined with one or more compounds pounds of formula (I) and (A1-A4) with one or more inter selected from the group consisting of non-nucleoside inhibi ferons, ribavirin or its analogs, HCV NS3 protease inhibitors, tors of HCV NS5B polymerase (ABT-072 and ABT-333), alpha-glucosidase 1 inhibitors, hepatoprotectants, nucleoside HCVNS5A inhibitors (ACH-3102 and ACH-2928) and HCV or nucleotide inhibitors of HCV NS5B polymerase, non 45 NS3 protease inhibitors (ABT-450 and ACH-1625). nucleoside inhibitors of HCV NS5B polymerase, HCV In yet another embodiment, the present application dis NS5A inhibitors, TLR-7 agonists, cyclophillin inhibitors, closes pharmaceutical compositions comprising a compound HCV IRES inhibitors, pharmacokinetic enhancers, and other as described herein, or a pharmaceutically acceptable salt, drugs or therapeutic agents for treating HCV. solvate, and/or ester thereof, in combination with at least one More specifically, one or more compounds as described 50 additional therapeutic agent, and a pharmaceutically accept herein may be combined with one or more compounds able carrier or excipient. selected from the group consisting of: According to one embodiment, the therapeutic agent used 1) , e.g., pegylated rIFN-alpha2b (PEG-Intron), in combination with the compound as described herein can be pegylated rIFN-alpha2a (Pegasys), rIFN-alpha2b (Intron A), any agent having a therapeutic effect when used in combina rIFN-alpha 2a (Roferon-A), interferon alpha (MOR-22, 55 tion with the compound as described herein. For example, the OPC-18, Alfaferone, Alfanative, Multiferon, subalin), inter therapeutic agent used in combination with the compound as feron alfacon-1 (Infergen), interferon alpha-nil (Wellferon), described herein can be interferons, ribavirin analogs, NS3 interferon alpha-n3 (Alferon), interferon-beta (Avonex, protease inhibitors, NS5b polymerase inhibitors, alpha-glu DL-8234), interferon-omega (omega DUROS, Biomed 510), cosidase 1 inhibitors, hepatoprotectants, non-nucleoside albinterferon alpha-2b (Albuferon), IFN alpha-2b XL, BLX 60 inhibitors of HCV, and other drugs for treating HCV. 883 (Locteron), DA-3021, glycosylated interferon alpha-2b In another embodiment, the present application provides (AVI-005), PEG-Infergen, PEGylated interferon lambda-1 pharmaceutical compositions comprising a compound of for (PEGylated IL-29), and belerofon; 2) ribavirin and its ana mula (I), or a pharmaceutically acceptable salt, Solvate, and/ logs, e.g., ribavirin (Rebetol, Copegus), and (Vira or ester thereof, in combination with at least one additional midine); 65 therapeutic agent selected from the group consisting of pegy 3) HCV NS3 protease inhibitors, e.g., (SCH lated rIFN-alpha2b, pegylated riFN-alpha2a, riFN-alpha2b, 503034, SCH-7), (VX-950), TMC435350, IFN alpha-2b XL, rIFN-alpha2a, consensus IFN alpha, infer US 8,940,718 B2 21 22 gen, rebif, locteron, AVI-005, PEG-infergen, pegylated IFN HIV, for example HIV protease inhibiting compounds, non beta, oral interferon alpha, feron, reaferon, intermax alpha, nucleoside inhibitors of HIV , HIV r-IFN-beta, infergen-actimmune, IFN-omega with DUROS, nucleoside inhibitors of reverse transcriptase, HIV nucleotide albuferon, rebetol, copegus, levovirin, VX-497, viramidine inhibitors of reverse transcriptase, HIV integrase inhibitors, (taribavirin), A-831, A-689, NM-283, valopicitabine, R1626, inhibitors, CXCR4 inhibitors, gp120 inhibitors, CCR5 PSI-6130 (R1656), HCV-796, BILB 1941, MK-0608, inhibitors, interferons, ribavirin analogs, NS3 protease NM-107, R7128, VCH-759, PF-868554, GSK625433, XTL inhibitors, NS5b polymerase inhibitors, alpha-glucosidase 1 2125, SCH-503034 (SCH-7), VX-950 (Telaprevir), ITMN inhibitors, hepatoprotectants, non-nucleoside inhibitors of 191, and BILN-2065, MX-3253 (celgosivir), UT-231B, IDN HCV, and other drugs for treating HCV. 6556, ME 3738, MitoC), and LB-84451, benzimidazole 10 More specifically, one or more compounds of formula (I) derivatives, benzo-1,2,4-thiadiazine derivatives, and pheny or (A1)-(A4) may be combined with one or more compounds lalanine derivatives, Zadaxin, nitazoxanide (alinea), BIVN selected from the group consisting of 1) HIV protease inhibi 401 (virostat), DEBIO-025, VGX-410C, EMZ-702, AVI tors, e.g., , , , , 4065, bavituximab, oglufanide, PYN-17, KPE02003002, , , lopinavir-i-ritonavir, , actilon (CPG-10101), KRN-7000, civacir, GI-5005, ANA 15 , , , , TMC-126. 975 (isatoribine), XTL-6865, ANA971, NOV-205, tarvacin, TMC-114, (DMP-450), JE-2147 (AG1776), EHC-18, and NIM811 and a pharmaceutically acceptable AG 1859, DG35, L-756423, ROO334649, KNI-272, DPC carrier or excipient. 681, DPC-684, and GW640385X, DG17, PPL-100, 2) a HIV In yet another embodiment, the present application pro non-nucleoside inhibitor of reverse transcriptase, e.g., vides a combination pharmaceutical agent comprising: , , delaviridine, , , a) a first pharmaceutical composition comprising a com (+) , , GW5634, DPC-083, DPC-961, pound of formula (I), or a pharmaceutically acceptable salt, DPC-963, MIV-150, and TMC-120, TMC-278 (), solvate, or ester thereof, and efavirenz, BILR 355 BS, VRX 840773, UK-453,061, b) a second pharmaceutical composition comprising at RDEA806, 3) a HIV nucleoside inhibitor of reverse tran least one additional therapeutic agent selected from the group 25 Scriptase, e.g., , emitricitabine, , stavu consisting of HIV protease inhibiting compounds, HIV non dine, , , , , elvucit nucleoside inhibitors of reverse transcriptase, HIV nucleo abine, , MIV-210, (-t-FTC), D-da-FC, side inhibitors of reverse transcriptase, HIV nucleotide , phosphazide, fozivudine tidoxil, fosalvudine inhibitors of reverse transcriptase, HIV integrase inhibitors, tidoxil, apricitibine (AVX754), amdoxovir, KP-1461, aba gp41 inhibitors, CXCR4 inhibitors, gp120 inhibitors, CCR5 30 cavir-lamivudine, abacavir-lamivudine--zidovudine, inhibitors, interferons, ribavirin analogs, NS3 protease zidovudine--lamivudine, 4) a HIV nucleotide inhibitor of inhibitors, alpha-glucosidase 1 inhibitors, hepatoprotectants, reverse transcriptase, e.g., tenofovir, non-nucleoside inhibitors of HCV, and other drugs for treat fumarate--emitricitabine, tenofovir disoproxil fumarate-- ing HCV, and combinations thereof. emtricitabine--efavirenz, and . 5) a HIV integrase In another embodiment is provided a pharmaceutical com 35 inhibitor, e.g., curcumin, derivatives of curcumin, chicoric position comprising a compound of formula (I) as described acid, derivatives of chicoric acid, 3.5-dicaffeoylquinic acid, herein and a nucleoside or nucleotide inhibitors of HCV derivatives of 3,5-dicaffeoylquinic acid, aurintricarboxylic NS5B polymerase and optionally an interferon or ribavirin. In acid, derivatives of aurintricarboxylic acid, caffeic acid phen one embodiment, the compound is methyl (2S)-1-(2S,5S)- ethyl ester, derivatives of caffeic acid phenethyl ester, tyr 2-(9-2-(2S,4S)-1-(2R)-2-[(methoxycarbonyl)amino-2- 40 phostin, derivatives of tyrphostin, quercetin, derivatives of phenylacetyl)-4-(methoxymethyl)pyrrolidin-2-yl)-1H-imi quercetin, S-1360, Zintevir (AR-177), L-870812, and dazol-5-yl)-1,11-dihydroisochromeno4',3':6.7naphtho1, L-870810, MK-0518 (), BMS-707035, MK-2048, 2-diimidazol-2-yl)-5-methylpyrrolidin-1-yl)-3-methyl-1- BA-011, BMS-538158, GSK364735C, 6) a gp41 inhibitor, Oxobutan-2-yl)carbamate having the formula: e.g., , sifuvirtide, FB006M, TRI-1144, SPC3,

and the inhibitor is Sofosbuvir. DES6, Locus gp41, CovX, and REP 9, 7) a CXCR4 inhibitor, Combinations of the compounds of formula I and addi e.g., AMD-070, 8) an , e.g., SP01A, TNX-355, tional active therapeutic agents may be selected to treat 9) a gp120 inhibitor, e.g., BMS-488043 and Block Aide/CR, patients infected with HCV and other conditions such as HIV 65 10) a G6PD and NADH-oxidase inhibitor, e.g., immunitin, . Accordingly, the compounds of formula I may be 11) a CCR5 inhibitor, e.g., , , INCB9471, combined with one or more compounds useful in treating PRO-140, INCB15050, PF-232798, CCR5 mAb004, and US 8,940,718 B2 23 24 . 12) an interferon, e.g., pegylated rFN-alpha 2b, Methods for determining stability of compounds in surro pegylated rFN-alpha 2a, rIFN-alpha2b. IFN alpha-2b XL, gate gastrointestinal secretions are known. rIFN-alpha2a, consensus IFN alpha, infergen, rebif, locteron, AVI-005, PEG-infergen, pegylated IFN-beta, oral interferon Exemplary Methods of Making the Compounds alpha, feron, reaferon, intermax alpha, r-IFN-beta, infergen-- 5 actimmune. IFN-omega with DUROS, and albuferon, 13) The disclosure also relates to methods of making the com ribavirin analogs, e.g., rebetol, copegus, levovirin, VX-497. positions of the disclosure. The compositions are prepared by and viramidine (taribavirin), 14) NS5a inhibitors, e.g., A-831, any of the applicable techniques of organic synthesis. Many A-689, and BMS-790052, 15) NS5b polymerase inhibitors, Such techniques are well known in the art. However, many of 10 the known techniques are elaborated in Compendium of e.g., NM-283, valopicitabine, R1626, PSI-6130 (R1656), Organic Synthetic Methods (John Wiley & Sons, New York), HCV-796, BILB 1941, MK-0608, NM-107, R7128, VCH Vol. 1, Ian T. Harrison and Shuyen Harrison, 1971; Vol. 2, Ian 759, PF-868554, GSK625433, and XTL-2125, 16) NS3 pro T. Harrison and Shuyen Harrison, 1974; Vol. 3. Louis S. tease inhibitors, e.g., SCH-503034 (SCH-7), VX-950 (Tela Hegedus and Leroy Wade, 1977; Vol. 4, Leroy G. Wade, Jr., previr), ITMN-191, and BILN-2065, 17) alpha-glucosidase 1 15 1980; Vol. 5, Leroy G. Wade, Jr., 1984; and Vol. 6, Michael B. inhibitors, e.g., MX-3253 (celgosivir) and UT-231B, 18) Smith; as well as March, J., Advanced Organic Chemistry, hepatoprotectants, e.g., IDN-6556, ME 3738, MitoO, and Third Edition, (John Wiley & Sons, New York, 1985), Com LB-84451, 19) non-nucleoside inhibitors of HCV, e.g., ben prehensive Organic Synthesis. Selectivity, Strategy & Effi Zimidazole derivatives, benzo-1,2,4-thiadiazine derivatives, ciency in Modern Organic Chemistry. In 9Volumes, Barry M. and phenylalanine derivatives, 20) other drugs for treating Trost, Editor-in-Chief (Pergamon Press, New York, 1993 Hepatitis C, e.g., Zadaxin, nitazoxanide (alinea), BIVN-401 printing). Other methods suitable for preparing compounds (virostat), DEBIO-025, VGX-410C, EMZ-702, AVI 4065, of the disclosure are described in International Patent Appli bavituximab, oglufanide, PYN-17, KPE02003002, actilon cation Publication Number WO 2006/020276. (CPG-10101), KRN-7000, civacir, GI-5005, ANA-975 (isa A number of exemplary methods for the preparation of the toribine), XTL-6865, ANA 971, NOV-205, tarvacin, EHC 25 compositions of the disclosure are provided in the schemes 18, and NIM811, 20) pharmacokinetic enhancers, e.g., BAS and examples below. These methods are intended to illustrate 100 and SPI452, 21) RNAse H inhibitors, e.g., ODN-93 and the nature of Such preparations and are not intended to limit ODN-112, 22) other anti-HIV agents, e.g., VGV-1, PA-457 the scope of applicable methods. (), ampligen, HRG214, cytolin, polymun, VGX Generally, the reaction conditions such as temperature, 410, KD247, AMZ 0026, CYT 99007, A-221 HIV, BAY 30 reaction time, solvents, work-up procedures, and the like, will 50-4798, MDX010 (iplimumab), PBS119, ALG889, and be those common in the art for the particular reaction to be PA-1050040. performed. The cited reference material, together with mate It is contemplated that the second therapeutic agent will be rial cited therein, contains detailed descriptions of such con administered in a manner that is known in the art and the ditions. Typically the temperatures will be -100° C. to 200° dosage may be selected by someone of skill in the art. For 35 C., solvents will be aprotic or protic, and reaction times will example, the second agent may be administered in a dose be 10 seconds to 10 days. Work-up typically consists of from about 0.01 milligrams to about 2 grams per day. quenching any unreacted reagents followed by partition between a water/organic layer system (extraction) and sepa Metabolites of the Compounds rating the layer containing the product. 40 Oxidation and reduction reactions are typically carried out Also falling within the scope of this disclosure are the in at temperatures near room temperature (about 20° C.), vivo metabolic products of the compounds described herein. although for metal hydride reductions frequently the tem Such products may result for example from the oxidation, perature is reduced to 0°C. to -100°C., solvents are typically reduction, hydrolysis, amidation, esterification and the like of aprotic for reductions and may be either protic or aprotic for the administered compound, primarily due to enzymatic pro- 45 oxidations. Reaction times are adjusted to achieve desired cesses. Accordingly, the disclosure includes compounds pro conversions. duced by a process comprising contacting a compound of this Condensation reactions are typically carried out at tem disclosure with a mammal for a period of time sufficient to peratures near room temperature, although for non-equili yield a metabolic product thereof. Such products typically are brating, kinetically controlled condensations reduced tem identified by preparing a radiolabelled (e.g., C'' or H) com- 50 peratures (0°C. to -100°C.) are also common. Solvents can pound of the disclosure, administering it parenterally in a be either protic (common in equilibrating reactions) or apro detectable dose (e.g., greater than about 0.5 mg/kg) to an tic (common in kinetically controlled reactions). animal Such as rat, mouse, guinea pig, monkey, or to man, Standard synthetic techniques such as azeotropic removal allowing Sufficient time for metabolism to occur (typically of reaction by-products and use of anhydrous reaction condi about 30 seconds to 30 hours) and isolating its conversion 55 tions (e.g., inert gas environments) are common in the art and products from the urine, blood or other biological samples. will be applied when applicable. These products are easily isolated since they are labeled (oth The terms “treated”, “treating, “treatment, and the like, ers are isolated by the use of antibodies capable of binding when used in connection with a chemical synthetic operation, epitopes surviving in the metabolite). The metabolite struc mean contacting, mixing, reacting, allowing to react, bring tures are determined in conventional fashion, e.g., by MS or 60 ing into contact, and other terms common in the art for indi NMR analysis. In general, analysis of metabolites is done in cating that one or more chemical entities is treated in Such a the same way as conventional studies well manner as to convert it to one or more other chemical entities. known to those skilled in the art. The conversion products, so This means that “treating compound one with compound long as they are not otherwise found in Vivo, are useful in two' is synonymous with “allowing compound one to react diagnostic assays for therapeutic dosing of the compounds of 65 with compound two', 'contacting compound one with com the disclosure even if they possess no HCV-inhibitory activity pound two”, “reacting compound one with compound two'. of their own. and other expressions common in the art of organic synthesis US 8,940,718 B2 25 26 for reasonably indicating that compound one was “treated'. pounds with chiral derivatizing reagents, separation of the “reacted”, “allowed to react', etc., with compound two. For diastereomers, and conversion to the pure stereoisomers, and example, treating indicates the reasonable and usual manner (3) separation of the substantially pure or enriched stereoiso in which organic chemicals are allowed to react. Normal mers directly under chiral conditions. concentrations (0.01M to 10M, typically 0.1M to 1M), tem Under method (1), diastereomeric salts can be formed by peratures (-100° C. to 250° C., typically -78° C. to 150° C., reaction of enantiomerically pure chiral bases such as bru more typically -78°C. to 100° C., still more typically 0°C. to cine, quinine, ephedrine, Strychnine, C.-methyl-3-phenyl 100° C.), reaction vessels (typically glass, plastic, metal), ethylamine (amphetamine), and the like with asymmetric Solvents, pressures, atmospheres (typically airfor oxygen and compounds bearing acidic functionality. Such as carboxylic water insensitive reactions or nitrogen or argon for oxygen or 10 water sensitive), etc., are intended unless otherwise indicated. acid and Sulfonic acid. The diastereomeric salts may be The knowledge of similar reactions known in the art of induced to separate by fractional crystallization or ionic chro organic synthesis is used in selecting the conditions and appa matography. For separation of the optical isomers of amino ratus for “treating in a given process. In particular, one of compounds, addition of chiral carboxylic or Sulfonic acids, ordinary skill in the art of organic synthesis selects conditions 15 Such as camphorsulfonic acid, tartaric acid, mandelic acid, or and apparatus reasonably expected to Successfully carry out lactic acid can result information of the diastereomeric salts. the chemical reactions of the described processes based on Alternatively, by method (2), the substrate to be resolved is the knowledge in the art. reacted with one enantiomer of a chiral compound to form a Modifications of each of the exemplary schemes and in the diastereomeric pair (Eliel, E. and Wilen, S. (1994) Stere Examples (hereafter “exemplary schemes”) leads to various ochemistry of Organic Compounds, John Wiley & Sons, Inc., analogs of the specific exemplary materials produced. The p. 322). Diastereomeric compounds can be formed by react above-cited citations describing Suitable methods of organic ing asymmetric compounds with enantiomerically pure synthesis are applicable to such modifications. chiral derivatizing reagents, such as menthyl derivatives, fol In each of the exemplary Schemes it may be advantageous lowed by separation of the diastereomers and hydrolysis to to separate reaction products from one another and/or from 25 yield the free, enantiomerically enriched substrate. A method starting materials. The desired products of each step or series of determining optical purity involves making chiral esters, of steps is separated and/or purified (hereinafter separated) to Such as a menthyl ester, e.g., (-) menthyl chloroformate in the the desired degree of homogeneity by the techniques com presence of base, or Mosher ester, C.-methoxy-O-(trifluorom mon in the art. Typically such separations involve multiphase ethyl)phenyl acetate (Jacob III. (1982) J. Org. Chem. extraction, crystallization from a solvent or solvent mixture, 30 distillation, Sublimation, or chromatography. Chromatogra 47:4165), of the racemic mixture, and analyzing the NMR phy can involve any number of methods including, for spectrum for the presence of the two atropisomeric diastere example: reverse-phase and normal phase; size exclusion; ion omers. Stable diastereomers of atropisomeric compounds can exchange; high, medium, and low pressure liquid chromatog be separated and isolated by normal- and reverse-phase chro raphy methods and apparatus; Small scale analytical; simu 35 matography following methods for separation of atropiso lated moving bed (SMB) and preparative thin or thick layer meric naphthyl-isoquinolines (Hoye, T., WO 96/15111). By chromatography, as well as techniques of Small scale thin method (3), a racemic mixture of two enantiomers can be layer and flash chromatography. separated by chromatography using a chiral stationary phase Another class of separation methods involves treatment of (Chiral Liquid Chromatography (1989) W. J. Lough, Ed. a mixture with a reagent selected to bind to or render other 40 Chapman and Hall, New York: Okamoto, (1990).J. of Chro wise separable a desired product, unreacted starting material, matogr: 513:375-378). Enriched or purified enantiomers can reaction by product, or the like. Such reagents include adsor be distinguished by methods used to distinguish other chiral bents or absorbents such as activated carbon, molecular molecules with asymmetric carbon atoms, such as optical sieves, ion exchange media, or the like. Alternatively, the rotation and circular dichroism. reagents can be acids in the case of a basic material, bases in 45 the case of an acidic material, binding reagents such as anti Schemes and Examples bodies, binding proteins, selective chelators such as crown ethers, liquid/liquidion extraction reagents (LIX), or the like. General aspects of these exemplary methods are described Selection of appropriate methods of separation depends on below and in the Examples. Each of the products of the the nature of the materials involved. For example, boiling 50 following processes is optionally separated, isolated, and/or point, and molecular weight in distillation and Sublimation, purified prior to its use in Subsequent processes. presence or absence of polar functional groups in chromatog A number of exemplary methods for the preparation of raphy, stability of materials in acidic and basic media in compounds of the disclosure are provided herein, for multiphase extraction, and the like. One skilled in the art will example, in the Examples below. These methods are intended apply techniques most likely to achieve the desired separa 55 tion. to illustrate the nature of Such preparations and are not A single stereoisomer, e.g., an enantiomer, Substantially intended to limit the scope of applicable methods. Certain free of its stereoisomer may be obtained by resolution of the compounds of the disclosure can be used as intermediates for racemic mixture using a method Such as formation of diaste the preparation of other compounds of the disclosure. In the reomers using optically active resolving agents (Stereochem 60 exemplary methods described herein, the fragment E-V— istry of Carbon Compounds, (1962) by E. L. Eliel, McGraw can also be written as R9-. PG represents a protecting group Hill; Lochmuller, C. H., (1975).J. Chromatogr., 113, 3) 283 common for the given functional group that it is attached. The 302). Racemic mixtures of chiral compounds of the disclo installation and removal of the protecting group can be Sure can be separated and isolated by any suitable method, accomplished using standard techniques, such as those including: (1) formation of ionic, diastereomeric salts with 65 described in Wuts, P. G. M., Greene, T. Protective Groups in chiral compounds and separation by fractional crystallization Organic Synthesis, 4th ed.: John Wiley & Sons, Inc.: Hobo or other methods, (2) formation of diastereomeric com ken, N.J., 2007. US 8,940,718 B2 27 28 -continued Scheme 1. Representative synthesis of E-W E-V-C(=O)-P-W-P-C(=O)-V-E Y O N

N-- N la o 10 W-E 2e

Scheme 2 shows a general synthesis of an E-V C(=O)— 15 P W P C(=O) V-E molecule of the disclosure wherein, for illustrative purposes, P is pyrrolidine. Coupling of amine 2a with acid 2b is accomplished using a peptide coupling reagent (e.g. HATU) to afford 2c. Alternatively, amine 2d is coupled with two equivalents of 2b under similar conditions to provide 2e.

Scheme 6. Representative synthesis of R-V-C(=O)-P-R2 25 p-v-c-o-r-w-O -- N H 6a 30 Scheme 1 shows a general synthesis of an E-V C(=O)— HO P W P C(=O) V-E molecule of the disclosure W-NH-PG --> wherein, for illustrative purposes, E is methoxycarbony lamino. The treatment of either 1a or 1c with one or two O 6b equivalents respectively of methyl chloroformate under basic 35 conditions (e.g. sodium hydroxide) provides the molecule 1b or 1d. p-v-c-o-r-w-O N

Scheme 2. Representative synthesis of 40 O E-V-C(=O)-P-W-P-C(=O)-V-E W-NH-PG 6c p-v-c-d-e-w-O -- 45 re-in-y-c-o-r-w-O -- N N H 2a 6d HO HO X-v-E Her 50 O X-v-E Her O 2b 6e

p-v-c-o-r-w-O 55 N re-in-y-c-o-r-w-O - N W-E o W-E 60 6f HO re-in-y-c-o-r-w-O -- O)—w-ON + 2 X-v- Hs N H O H 65 2d 2b 6d US 8,940,718 B2 30 -continued poses, P is pyrrolidine, R' is a generic group that is depicted HO as either-E or a amino protecting group, and R is a generic group that is depicted as —W P C(=O) V-E, X-v-N-PG -- W. P. C(=O) V. NH PG, W P NH PG, or O —W NH-PG. Coupling of amine 6a (or 6d, 6h, 6k) with acid 6b or 6e is accomplished using a peptide coupling reagent (e.g. HATU) to afford 6c (or 6f 6g, 6i, 6, 61, 6m) re-in-y-c-o-r-w-O respectively. N 10 O W-NH-PG Scheme 7. Representative synthesis of E-V-C(=O)-R 6g O HO re-in-r-w-O -- X-v-E 15 n NH O

- N W-E 25 6i HO re-in--w-O -- W-NH-PG NH O 30 6h 6b

re-in-e-w-ON - 35 W-NH-PG

40

45

50

55

60

W-NH-PH

65 Scheme 6 shows a general synthesis of an R' V C (=O) P R intermediate wherein, for illustrative pur US 8,940,718 B2 31 32 -continued -continued O X-Ni-v-c=o-o-re HO O X--Pg HipO

Scheme 7 shows a general synthesis of an E-V C(=O)— R' intermediate wherein, for illustrative purposes, E is meth 10 oxycarbonylamino and R' is a generic group that is depicted as either P W P C(=O) V. NH PG, P W P PG, P W PG, P PG, or - O PG. Treatment of 7a (or 7c, 7e, 7g,7i) with methyl chloroformate under basic Br conditions (e.g. sodium hydroxide) provides the molecule 7b 15 (or 7d, 7f, 7h, 7). X-r-re Scheme 9. Representative synthesis of R-P-R’ HO NH2 P-C(=O)-V-E

--> Br Br NH2 9b 25

9a NH2 30 Scheme 9 shows a general synthesis of an R P R Br NH -- intermediate wherein, for illustrative purposes, R' is X-r-c=o-v- —C(=O) V-E or a protecting group and R is a substituted O benzimidazole. The formation of the benzimidazole is 9c 35 accomplished by coupling the acid 9b or 9e with an arylamine 9a, using a peptide coupling reagent such as HATU, to afford 9c or 9f. Cyclization of the amide in the presence of an acid ----- (such as acetic acid) affords the benzimidazole containing Br N molecule 9d or 9g. 40 The formation of multiple benzimidazoles is performed in 9d the same manner, starting with a bis-diamine to provide the corresponding bis-benzimidazole.

Scheme 20. Representative synthesis of R-P-W-P-R2 Br O

- HO -as- O -e-

C Br Br

C 20a 2Ob 20c O O

O O Br

C C 20d 20e US 8,940,718 B2 33 34 -continued

P-PG O 1-( O O P-PG O N O O He- CO His

C C 2Of 20g/h P-PG O -( PG-P

CO 20k X

O

20i, O H N P-PG usN1 N I r

20m

Scheme 20 shows a general synthesis of an R'—P W P R intermediate of the disclosure wherein, for illustrative Scheme 21. Representative synthesis of R-P-W-P-R purposes, RandR are independent protecting groups and W 40 O is a two aromatic ring unit constructed via a transition metal O mediated cyclization. Alkylation of phenol 20b with an alkyl bromide, such as 20a, provides the ether 20c. Cyclization of -e- the aromatic rings in the presence of a palladium catalyst provides the compound 20d. Treatment of 20d with Cubr, provides the C-haloketone 20e, which provides 20f upon addition of an acid under basic conditions (e.g. EtsN). Reac C tion of 20f with an amine or amine salt (e.g. ammonium 20d O acetate) affords the imidazole containing molecule 20g. Oxi- 50 O dation of 20g, 20i, or 201 can be accomplished by heating in the presence of MnO, to provide 20h, 20, or 20m, respec- O -- tively. Conversion of 20g or 20h with a palladium catalyst, Such as Pd dba and X-Phos, and a boron Source Such as bis(pinacolato) diboron provides the boronic ester 20i or 20. ss S The boronic ester is coupled with an appropriate coupling partner (e.g. 20k) using a palladium catalyst, such as 21a O Pd(PPh) or PdCl(dppf), to afford 201 or 20m. For each O transition metal mediated cross-coupling reaction, the roles of the nucleophile and electrophile can be reversed to provide 60 the same coupling product. Other transition metal mediated Br -e- cross couplings that enable the construction of W. but employ alternative coupling partners and reagents, include, but are not limited to, the Negishi, Kumada, Stille, and Ullman cou plings. For the preparation of alternate two aromatic ring 65 O containing W groups, this general scheme can be applied through the appropriate choice of the starting reagents. 21b US 8,940,718 B2 36 -continued -continued O O O H O N1 N --- ls -e- 5 PG-P O --l \ / \-( ) I 21 fg 10 O 21c Scheme 21 shows a general synthesis of an R'—P W O P R intermediate of the disclosure wherein, for illustrative O Br purposes, RandR are independent protecting groups and W O 15 is a two aromatic ring unit constructed via a transition metal mediated cyclization. Treatment of 20d with an activated --> vinyl reagent (e.g. potassium vinyltrifluoroborate) in the presence of a palladium catalyst (e.g. palladium acetate and 2O S-Phos) provides the vinyl compound 21a. Conversion to the O corresponding C-halo ketone can be accomplished by bromi 21d nation with N-bromosuccinimide, followed by oxidation with MnO. Displacement of the O.-halo ketone proceeds by the addition of an acid under basic conditions (e.g. EtN). 25 Bromination of 21c proceeds upon treatment with pyridinium tribromide, and is followed by the addition of a second acid under basic conditions to provide the diester 21e. Reaction of 21e with an amine or amine Salt (e.g. ammonium acetate) 30 affords the imidazole containing molecule 21f. Oxidation of 21e 21f can be accomplished in the presence of MnO, to provide 21g.

Scheme 22. Representative synthesis of E-V-C(=O)-P-W-P-R O O

O O O He- ls He Br E-V-C(=O)-P O

O O 21b 22a

O

O Br O ls -as E-V-C(=O)-P O

O 22b US 8,940,718 B2 37 38 -continued

O

O O P-PG O r ls O -e- E-V-C(=O)-P O

O 22c

O

H o ----- ( ) N 22dfe

Scheme 22 shows a general synthesis of an E-V C treatment with pyridinium tribromide, and is followed by the (=O)—P W P R intermediate of the disclosure addition of a second acid under basic conditions to provide wherein, for illustrative purposes, R is a protecting group and the diester 22c. Reaction of 22c with an amine or amine salt W is a two aromatic ring unit. Displacement of the C-halo (e.g. ammonium acetate) affords the imidazole containing ketone 21b proceeds by the addition of an acid under basic molecule 22d. Oxidation of 22d can be accomplished in the conditions (e.g. EtsN). Bromination of 22a proceeds upon presence of MnO, to provide 22e.

Scheme 23. Representative synthesis of R-P-W-P-C(=O)-V-E O

O B r O ls, -e-

O 21d O

O O O r O PG-P ls,

O 23a O

H Y. u Wu ul-N N s---| W-E

23 bic US 8,940,718 B2 39 40 Scheme 23 shows a general synthesis of an E-V C -continued (=O)—P W P R intermediate of the disclosure wherein, for illustrative purposes, R is a protecting group and PG-P W is a two aromatic ring unit. Displacement of the C-halo ketone 21d proceeds by the addition of an acid under basic conditions (e.g. EtsN). Reaction of 23a with an amine or amine salt (e.g. ammonium acetate) affords the imidazole containing molecule 23b. Oxidation of 23b can be accom plished in the presence of MnO, to provide 23c. 10 ----- X Scheme 25. Representative synthesis of E-V-C(=O)-P-W-P-C(=O)-V-E 15 Scheme 26 shows a general synthesis of an E-V C O (=O) P R and an R P R molecule of the disclosure wherein, for illustrative purposes R is a haloimidazole. Treat N-- ment of the aldehyde 26a with glyoxal, in the presence of ammonium hydroxide provides the imidazole 26b. Treatment with either N-bromosuccinamide or iodine provides the cor responding haloimidazole 26c and 26d respectively. Separa tion from the corresponding bis-halogenated compound can be accomplished by preparative HPLC chromatography. The conversion of the bis-haloimidazole to the mono-haloimida 25 25b Zole can also be accomplished upon heating in the presence of sodium sulfite. Further functionalization of the P group can be accomplished upon removal of the protecting group and cou nullsC pling with an appropriate acid (E-V C(=O)—OH). 30 Scheme 27. Representative synthesis of R-P-W-P-R O

35

25

Scheme 25 shows a general synthesis of an E-V C 40 (=O) P W P C(=O) V-E molecule of the disclo sure wherein, for illustrative purposes, E is ethylcarbony lamino. The treatment of either 25a or 25c with one or two equivalents respectively of propionyl chloride under basic 21b conditions (e.g. sodium hydroxide) provides the molecule 45 25b or 25d.

Scheme 26. Representative syntheses of E-V-C(=O-P-R and R-P-R 50

PG-P PG-P O -- N y/ -- 55 27a

26a. 26b ---X 26c ------X 65 26e 21d US 8,940,718 B2 41 42 -continued Scheme 27 shows an alternate general synthesis of an O O P-PG R P W P R intermediate of the invention wherein, O ls —- for illustrative purposes, R' and Rare independent protect PG-P O O ing groups and W is a two aromatic ring unit constructed via a transition metal mediated cyclization. Bromination of 21b O 10 with a brominating agent (i.e. pyridinium tribromide) pro 21e O vides the dibromide 27a. Displacement of the primary bro H N P-PG mide then proceeds by the addition of an acid under basic Y-K)-( )-(? conditions (e.g. KCO) to provide 21d. Conversion to 21 for H 21g can be accomplished following methods described in 21 fg Scheme 21.

Scheme 28. Representati ve synthesis of E-V-C(=O)-P-W-P-R O O

O O Br

Br Br

O O 21b 27a

O

O Br O

lsO -e-

O 22b O

O O P-PG O ls O He E-V-C(=O)-P O

O J)-Rs.( ) C. r. 22dfe US 8,940,718 B2 43 44 Scheme 28 shows an alternate general synthesis of an (e.g. KCO) to provide 22d. Conversion to 22d or 22e can be E-V C(=O) P W P R intermediate of the invention accomplished following methods described in Scheme 22. wherein, for illustrative purposes, R is a protecting group and W is a two aromatic ring unit. Bromination of 21b with a Specific Embodiments brominating agent (i.e. pyridinium tribromide) provides the dibromide 27a. Displacement of the primary bromide then In one embodiment the disclosure provides a compound proceeds by the addition of an acid under basic conditions which has formula:

(A1)

Ela-Vila-C(=O) Pla

(A2) Pll-C(=O)-V-El

(A3) Plb-C(=O)-V-El O Ela-Vila-C(=O) Pla

(A4)

Ela-Vila-C(=O) Pla

wherein the imidazole ring shown informula A1, A2, A3, and A4 is optionally Substituted with one or more groups inde 45 pendently selected from halo, haloalkyl, cyano, or alkyl; or a pharmaceutically acceptable salt or prodrug thereof. In one embodiment the disclosure provides a compound which has formula:

(A2)

O Ela-Vila-C(=O) Pla

(A4)

Ela-Vila-C(=O) Pla US 8,940,718 B2 45 46 wherein the imidazole ring shown in formula A2 and A4 is -continued optionally Substituted with one or more groups independently selected from halo, haloalkyl, cyano, or alkyl; N or a pharmaceutically acceptable salt or prodrug thereof. In one embodiment at least one of E' and E' is N(H) (alkoxycarbonyl). In one embodiment at least one of E' and E' is N(H) C(=O)CMe. In one embodiment both of E" and E' are N(H)C(=O) 10 In one embodiment one of P' and P' is: OMe. In one embodiment at least one of E" and E' is N(H) (cycloalkylcarbonyl) or N(H)(cycloalkyloxycarbonyl). N In one embodiment at least one of E' and E' is cyclopro pylcarbonylamino, cyclobutylcarbonylamino, cyclopropy s loxycarbonylamino or cyclobutyloxycarbonylamino. In one embodiment E" and E' are each independently selected from cyclopropylcarbonylamino, cyclobutylcarbo 1. O nylamino, cyclopropyloxycarbonylamino or methoxycarbo nylamino. In one embodiment at least one of V' and V' is: and the other of P' and P' is:

25 N

30

In one embodiment E-V' taken together are R' or In one embodiment one of P'la and P'lb is::. wherein E-V' taken together are R'. In one embodiment at least one of P' and P' is selected 3s from: N s

O 40 1. O

and the other of P' and P' is: 1. opo NO N s s

In one embodiment at least one of P' and P' is: In one embodiment P' and P' are each independently 55 selected from: N r

s - s - - - 19 F O O 65 r 1. US 8,940,718 B2 47 48 In one embodiment at least one of P' and P' is: -continued x r x-r

In one embodiment at least one of V' C(=O)— 15 OMe OMe P' and P' C(=O) V is: O

25 OMe O O

N O s

1. 35 O In one embodiment at least one of V' C(=O)— P' and P' C(=O) V is: In one embodiment both of - V' C(=O) P' and 40 —P' C(=O) V' are independently selected from:

x -x

65 O US 8,940,718 B2 49 50 -continued -continued O

O O 5 OMe v.N-3, v. NS-X. O O v N 10 S-X. SSX 19 O Me 15 19

v v. N OMe OMe s O SA X O O v v. 25 N-9,s O NY,

Os 30 1.

and the other of and Plb C 1. V C(O) P' 35 (=O) V is: In one embodiment one of V' C(=O)—P'— and P' C(=O) V' is: 40

45

C. C. 50

55 r 60 SSX 65 US 8,940,718 B2 51 52 -continued -continued

N 10

15 1.

In one embodiment the disclosure provides a compound of formula:

US 8,940,718 B2 53 54 -continued

S S

: V O O

s O O No ulN1 E O H O Y

N.Y.O N N ( ) ( ) 1. O 1. w NV N O --S H s N O

O /

O O O Fl (1)N ()-- N r r's

US 8,940,718 B2 57 58 -continued

or a pharmaceutically acceptable salt or prodrug thereof. 45 In one embodiment the disclosure provides a compound of formula:

or a pharmaceutically acceptable salt or prodrug thereof. In one embodiment the disclosure provides a compound of formula:

US 8,940,718 B2 61 62

-continued

or a pharmaceutically acceptable salt or prodrug thereof. -continued

The disclosure will now be illustrated by the following Moc Methoxycarbonyl non-limiting Examples. The following abbreviations are used MS Mass spectrometry throughout the specification, including the Examples. MTBE Methyl tert-butyl ether N Normal NADPH Nicotinamide dinucleotide phosphate (aq) Aqueous 25 NBS N-Bromosuccinimide (g) Gas NMM N-Methylmorpholine (s) Solid NMR Nuclear magnetic resonance o C. Degree Celsius on Ac Acetate Over night ACN Acetonitrile Papp Apparent permeability apprx Approximate 30 PBS Phosphate buffer system Bis-pinB/(Bpin), Bis(pinacolato)diboron (pinB)2 Po?tC Palladium on carbon BOCBOc ert-Butoxycarbonyl Ph Phenyl calcid Calculated Phg/PhOly Phenylglycine CCso 50% Cytotoxicity concentration Piy Pivalate COMU -(1-(Cyano-2-ethoxy-2- 35 oxoethylideneaminooxy)-dimethylaminomorpholino) Pro Proline uronium hexafluorophosphate pyr Pyridine Doublet dibenzalacetone C Quartet Dichloromethane qd Quartet of doublets Doublet of doublets Doublet of doublet of doublets 40 quant Quantitative DIPEADIEA N,N-Diisopropylethylamine quint Quintet DMA N,N-Dimethylacetamide rtRT Room temperature DMAP 4-Dimethylaminopyridine DME Dimethoxyethane Singlet DMEM Eagle's minimal essential medium SPhOS 2-Dicyclohexylphosphino-2',6'-dimethoxybiphenyl 45 DMF Dimethylformamide Triplet DMSOdmso Dimethylsulfoxide dppf 1'-bis(diphenylphosphanyl) ferrocene t-Bu tert-Butyl dt Doublet of triplets TEMPO (2.2,6,6-Tetramethyl-piperidin-1-yl)oxyl ECso Half maximal effective concentration ESI Electrospray ionization Tf Trifluoromethanesulfonate Et Ethyl 50 TFA Trifluoroacetic acid ext. External THF Tetrahydrofuran FBS Fetal bovine serum 9. Gram Thr Threonine HATU 2-(1H-7-AZabenzotriazol-1-yl)-1,1,3,3-tetramethyl TLC Thin layer chromatography uronium hexafluorophosphate Methanaminium tol. Toluene HPLC High performance liquid chromatography 55 hirih Hour Ultraviolet Hertz Wall Valine Coupling constant Liquid chromatography mass spectrometry wiv Weight to volume Molar ww Weight to weight Multiplet X-POSXPOS, 2-Dicyclohexylphosphino-2',4',6'- Mass to charge 60 Mass peak Xphos triisopropylbiphenyl Methyl 8 Chemical shift Milligram Megahertz 19. Microgram Minute |L Microliter Milliliter 65 Millimole

US 8,940,718 B2 65 66 -continued

1.HCl, EtOH, 60° C. 2.

O tert-butyl 2-9-(2-1-N-(methoxycarbonyl)valylpyrrollidin-2- 1. O ls o1 dihydroisochromeno4",3:6.7naphtho1,2-dimidazol-2- ylpyrrollidine-1-carboxylate O H

COMU, DIPEA, DMF, RT

1-(2-5-2-(1- {(methoxycarbonyl)amino (phenyl)acetylpyrrollidin -2-yl)-1,11 dihydroisochromeno4',3':6.7naphtho1,2- dimidazol-9-yl)-1H-imidazol-2-yl)pyrrollidin-1-yl)-3- methyl-1-oxobutan-2-ylcarbamic acid

7-(2-bromo-5-chlorobenzyloxy)-3,4-dihydronaph 11-dihydro-5H-dibenzoc.gchromen-8(9H)-one (14.4 g, thalen-1 (2H)-one 35 67% yield) as an off-white solid. To a stirred solution of 7-hydroxy-1-tetralone (13.9 g, 85.7 mmol) and 1-bromo-2-(bromomethyl)-4-chlorobenzene 9-bromo-3-chloro-10,11-dihydro-5H-dibenzoc.g (25.6 g. 90.0 mmol) in dimethylformamide (850 mL) was chromen-8(9H)-one added potassium carbonate (24g, 172 mmol). The reaction 40 was stirred under argon for 18 hours then diluted with ethyl To a mixture of 3-chloro-10,11-dihydro-5H-dibenzoc.g acetate (1 L). The organics were washed three times with chromen-8(9H)-one (14.8 g., 52 mmol) in chloroform (50 water and once with brine. The organic layer was then dried mL) and ethyl acetate (50 mL) was added copper(II) bromide with magnesium Sulfate, filtered and concentrated. To the (24.3 g, 104 mmol). The reaction was heated to 80°C. for 2 resulting oil was added methanol (500 mL) and the suspen 45 hours and then cooled to room temperature. The mixture was sion was agitated for thirty minutes. 7-(2-bromo-5-chlo diluted with dichloromethane and washed twice with a 5:1 robenzyloxy)-3,4-dihydronaphthalen-1 (2H)-one (27.8 g. Solution of saturated aqueous ammonium chloride and aque 89% yield) was isolated by filtration. ous ammonium hydroxide (-38%), and washed once with water. The organic layer was dried with magnesium sulfate, 3-chloro-10,11-dihydro-5H-dibenzoc.gchromen-8 50 filtered and concentrated to yield 9-bromo-3-chloro-10.11 (9H)-one dihydro-5H-dibenzoc.gchromen-8(9H)-one (18.5g, D-95% yield) with >95% purity. To a 1 L flask containing palladium(II) pivalate (1.18 g. 3.8 Note: This reaction is not always this clean. Sometimes mmol), tri(4-fluorophenyl)phosphine (1.20g, 3.8 mmol), piv there is over-bromination and sometimes there is significant alic acid (2.33 g, 22.8 mmol) and potassium carbonate (31.8 55 starting material. These impurities can be removed by flash g, 228 mmol) was added a solution of 7-(2-bromo-5-chlo column chromatography. robenzyloxy)-3,4-dihydronaphthalen-1 (2H)-one (27.8 g. 76.2 mmol) in dimethylacetamide (380 mL). The flask was evacuated and backfilled with argon 5 times and then stirred tert-Butyl 2-(9-chloro-1,4,5,11-tetrahydroisoch under argon at 60°C. for 24 hours. The reaction was cooled to 60 romeno4".3':6.7naphtho1,2-dimidazol-2-yl)pyrro room temperature and diluted with MTBE and water. The lidine-1-carboxylate resulting biphasic mixture was stirred for 3 hours and filtered through Celite, rinsing with MTBE. The organic layer of the To a solution of (1R)-2-(tert-butoxycarbonyl)cyclopentan filtrate was separated and then washed twice with water and ecarboxylic acid (10.17 g. 47.25 mmol) and 9-bromo-3- once with brine. The organics were then dried with magne 65 chloro-10,11-dihydro-6H-naphtho2.3-cchromen-8(9H)- sium sulfate, filtered, concentrated and purified by flash col one (5.7 mg, 15.7 mmol) in acetonitrile (50 mL) was added umn chromatography (Hexanes/DCM) to yield 3-chloro-10, diisopropylethylamine (11.11 mL. 64 mmol). The reaction US 8,940,718 B2 67 68 was stirred at 50° C. for 4 hours and was then diluted with mamide (6 mL) was added a solution of potassium carbonate ethyl acetate. The organics were washed with water and brine, (2M in water, 5 mL, 9.93 mmol). The resulting mixture was dried (MgSO) and concentrated. The resulting crude residue degassed and then heated to 85°C. under argon for 18 hours. was purified by flash chromatography to yield (2S)-1-tert After cooling to room temperature, the reaction was diluted butyl 2-(3-chloro-8-oxo-8,9,10,11-tetrahydro-5H-naphtho with ethyl acetate. The organics were washed with water and c.gchromen-9-yl)pyrrolidine-1,2-dicarboxylate (4.52 g, brine, dried (NaSO), and concentrated. The crude residue 58%). To a solution of (2S)-1-tert-butyl 2-(3-chloro-8-oxo-8, was purified by flash chromatography to yield tert-butyl 2-9- 9,10,11-tetrahydro-6H-naphtho2.3-cchromen-9-yl)pyrro (2-1-N-(methoxycarbonyl)valylpyrrolidin-2-yl)-1H-imi lidine-1,2-dicarboxylate (3.27 mg, 6.56 mmol) in a mixture dazol-5-yl)-1,11-dihydroisochromeno4.3':6.7naphtho1, of toluene (11 mL) and 2-methoxyethanol (0.7 mL) was 10 2-diimidazol-2-ylpyrrolidine-1-carboxylate (1.45 mg. added ammonium acetate (5.06 g. 65.6 mmol). The reaction 59%). LCMS-ESI: calculated for C41H47N7O6, 733.86; mixture was heated to 110° C. for 3 hours, cooled to room temperature and diluted with ethyl acetate. The organics were observed M+1": 734.87. washed with water and brine, dried (Na2SO), and concen trated. The crude residue was purified by flash chromatogra 15 1-(2-5-2-(1-(Methoxycarbonyl)amino (phenyl) phy to yield tert-butyl 2-(9-chloro-1,4,5,11-tetrahydroisoch acetylpyrrolidin-2-yl)-1,11-dihydroisochromeno4", romeno4".3':6.7naphtho1,2-dimidazol-2-yl)pyrrolidine 3':6.7naphtho1,2-dimidazol-9-yl)-1H-imidazol-2- 1-carboxylate (1.95 g, 61%). LCMS-ESI: calculated for ylpyrrolidin-1-yl)-3-methyl-1-oxobutan-2-yl) C27H28CINO3: 477.98; observed M+1: 478.47 tert carbamic acid Butyl 2-(9-chloro-1,11-dihydroisochromeno4".3':6.7naph tho1,2-dimidazol-2-yl)pyrrolidine-1-carboxylate To a solution of tert-butyl 2-(9-chloro-1,4,5,11-tetrahy A solution of tert-butyl 2-9-(2-14N-(methoxycarbonyl) droisochromeno4".3':6.7naphtho1,2-diimidazol-2-yl)pyr valylpyrrolidin-2-yl)-1H-imidazol-5-yl)-1,11-dihydroiso rolidine-1-carboxylate (1.9 g, 3.96 mmol) in dichlo chromeno4".3':6.7naphtho1,2-dimidazol-2-ylpyrroli romethane (35 mL) was added manganese(IV) oxide (17 g. 25 dine-1-carboxylate (462 mg, 0.63 mmol), ethanol (6 mL) and 198 mmol). The reaction mixture was stirred at room tem concentrated HCl (2 mL) was heated to 60°C. for 1 hour. The perature for 18 hours, diluted with ethyl acetate. The organics reaction was concentrated and the crude material dissolved in were washed with water and brine, dried (NaSO), and con DCM (6 mL). This solution was concentrated and to this centrated. The crude residue was purified by flash chroma material was added a solution of (R)-2-(methoxycarbony tography to yield tert-butyl 2-(9-chloro-1, 1'-dihydroisoch 30 lamino)-2-phenylacetic acid (172 mg, 0.82 mmol) and romeno4".3':6.7naphtho1,2-dimidazol-2-yl)pyrrolidine COMU (311 mg 0.73 mmol) in DMF (6 mL). To the resulting 1-carboxylate (1.52 g, 81%). LCMS-ESI: calculated for solution was added diisopropylethylamine (330 uI, 1.89 C27H26CIN3O3: 475.9; observed M+1": 476.45. mmol). After stirring for 18 hours at room temperature, the reaction was diluted with ethyl acetate, washed with water tert-Butyl 2-9-(4,4,5,5-tetramethyl-1,3,2-dioxaboro 35 and brine, dried (NaSO), concentrated and purified by pre lan-2-yl)-1,11-dihydroisochromeno4".3':6.7naphtho parative reverse phase HPLC (Gemini, 15 to 45% ACN/HO+ 1,2-dimidazol-2-ylpyrrolidine-1-carboxylate 0.1% TFA). The product fractions were lyophilized to give 1-(2-5-2-(1-(methoxycarbonyl)amino (phenyl) A degassed mixture of tert-butyl 2-(9-chloro-1,11-dihy acetylpyrrolidin-2-yl)-1,11-dihydroisochromeno4',3':6.7 droisochromeno4".3':6.7naphtho1,2-diimidazol-2-yl)pyr 40 rolidine-1-carboxylate (1.52g, 3.17 mmol), bis(pinacolato) naphtho1,2-diimidazol-9-yl)-1H-imidazol-2-ylpyrrolidin diboron (1.21 g, 4.75 mmol), potassium acetate (93.4 mg. 9.52 1-yl)-3-methyl-1-oxobutan-2-ylcarbamic acid (231 mg, mmol), tris(dibenzylideneacetone)palladium (116 mg, 0.13 45%). LCMS-ESI: calculated for C46H48N8O7: 824.92; mmol) and 2-dicyclohexylphosphino-2',4',6'-tri-1-propyl-1, observed M+1": 826.00. 1'-biphenyl (121 mg, 0.08 mmol) in 1,4-dioxane (16 mL) was 45 heated to 90° C. for 1.5 hours, cooled to room temperature Example LR and diluted with ethyl acetate. The organics were washed with water and brine, dried (NaSO), and concentrated. The crude residue was purified by flash chromatography to yield tert butyl 2-9-(4.4.5.5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1, 50 CN 11-dihydroisochromeno4".3':6.7naphtho1,2-diimidazol S 1) HCI, MeOH 2) Boc2O, NaHCO3 2-ylpyrrolidine-1-carboxylate (1.7 g.94%) He tert-Butyl 2-9-(2-1-N-(methoxycarbonyl)valyl MeOC'SO See pyrrolidin-2-yl)-1H-imidazol-5-yl)-1,11-dihydroiso 55 chromeno4".3':6.7naphtho1,2-dimidazol-2-yl) (2S4S)-1-tert-butyl 2-methyl 4 cyanopyrrollidine pyrrolidine-1-carboxylate 1,2-dicarboxylate To a solution of methyl (S)-1-((S)-2-(5-bromo-1H-imida COMe Zol-2-yl)pyrrolidin-1-yl)-3-methyl-1-oxobutan-2-ylcarbam 60 HipNaOH ate (1.48g, 3.97 mmol), tert-butyl 2-9-(4.4.5.5-tetramethyl H 1,3,2-dioxaborolan-2-yl)-1,11-dihydroisochromeno4".3':6, 7naphtho1,2-dimidazol-2-ylpyrrolidine-1-carboxylate MeOC See (1.88 g, 1.48 mmol), tetrakis(triphenyl phosphine)palladium (2S4S)-1-tert-butyl 2,4- (0) (191 mg 0.16 mmol) and dichloro1,1'-bis(diphenylphos 65 dimethyl pyrrollidine phino) ferrocenepalladium(II) (242 mg, 0.33 mmol) in a 1,2,4-tricarboxylate mixture of 1,2-dimethoxyethane (37.0 mL) and dimethylfor US 8,940,718 B2 70 -continued (2S,4S)-1-tert-butyl 2-methyl 4-(hydroxymethyl) CO2H pyrrolidine-1,2-dicarboxylate s 1) EtOCCI To a solution of (3S,5S)-1-(tert-butoxycarbonyl)-5-(meth HSO 2) NaBH4 oxycarbonyl)pyrrolidine-3-carboxylic acid (6.38 g, 23.3 MeOC See mmol) in THF (116 mL) at 0°C. (external temperature, ice (3S,5S)-1-(tert-butoxycarbonyl)- bath) was added EtN (4.9 mL, 35.0 mmol) and ethyl chlo 5-(methoxycarbonyl)pyrrollidine 3-carboxylic acid roformate (2.7 mL, 28.0 mmol). The resulting solution was HO 10 stirred at 0°C. for 45 min, during which time a white precipi tate forms. The reaction mixture was filtered through celite N MeI s (t-Bu)2pyr and concentrated. -e-AgOTf The crude intermediate was dissolved in THF (59 mL) and H cooled to 0°C. (external temperature, ice bath). NaBH (4.41 g, 116.7 mmol) in HO (59 mL) was slowly added and the MeOC See resulting solution was stirred at 0°C. for 2 h. The reaction (2S4S)-1-tert-butyl 2-methyl 4-(hydroxymethyl)pyrrollidine mixture was diluted with EtOAc and washed with HO. The 1,2-dicarboxylate aqueous layer was backextracted with EtOAc. The combined organic layers were dried over NaSO and concentrated. The - O\ - O\ crude oil was purified by silica gel chromatography (42% to s LiOH s 69% to 100% EtOAc/Hexanes) to provide (2S,4S)-1-tert -- butyl 2-methyl 4-(hydroxymethyl)pyrrolidine-1,2-dicar boxylate (3.63 g, 60%). 25 MeOC See HOC See (2S,4S)-1-tert-butyl 2-methyl 4-(methoxymethyl) (2S,4S)-1-tert-butyl 2-methyl (2S4S)-1-(tert 4-(methoxymethyl)pyrrollidine butoxycarbonyl)-4- pyrrolidine-1,2-dicarboxylate 1,2-dicarboxylate (methoxymethyl)pyrrollidine 2-carboxylic acid To a solution of (2S,4S)-1-tert-butyl 2-methyl 4-(hy 30 droxymethyl)pyrrolidine-1,2-dicarboxylate (2.57 g, 9.9 mmol) in CH2Cl (50 mL) was added AgOTf (4.07 g. 15.8 (2S,4S)-1-tert-butyl mmol) and 2,6-di-tert-butylpyridine (4.4 mL, 19.8 mmol). 2,4-dimethylpyrrolidine-1,2,4-tricarboxylate The reaction mixture was cooled to 0°C. (external tempera 35 ture, ice bath) and Mel (0.98 mL, 15.8 mmol) was slowly added. The resulting slurry was stirred at 0°C. for 1.5 hand To a solution of (2S,4S)-1-tert-butyl 2-methyl 4-cyanopy at room temperature for 1.5 h. The slurry was diluted with rrolidine-1,2-dicarboxylate (9.0g, 35.4 mmol) in MeCH (196 CH2Cl and filtered through celite. The filtrate was concen mL) was added HCl (4M in 1,4-dioxane, 100 mL, 403 mmol). trated to dryness, dissolved in EtO, and washed with HCl The solution was stirred at room temperature for 16 hand 40 (1N) and brine. The aqueous layers were backextracted with concentrated in vacuo. The crude intermediate was dissolved EtO and the combined organic layers were dried over in EtOAc (180 mL) and basified with aqueous bicarbonate NaSO and concentrated. The crude oil was purified by silica (sat.). Di-tert-butyl dicarbonate (8.5g, 38.9 mmol) was added gel chromatography (10% to 75% to 100% EtOAc/Hexanes) and the biphasic solution was stirred at room temperature for to provide (2S,4S)-1-tert-butyl 2-methyl 4-(methoxymethyl) 12 h. The layers were then separated and the aqueous layer 45 pyrrolidine-1,2-dicarboxylate (2.11 g, 78%). H-NMR: 400 was backextracted with EtOAc. The combined organic layers MHz, (CDC1) Ö: (mixture of rotamers, major reported) 4.20 were washed with brine, dried over NaSO, and concen trated. The crude oil was purified by silica gel chromatogra (t, 1H), 3.71 (s.3H), 3.67 (m, 1H), 3.34 (m, 2H), 3.30 (s.3H), phy (15% to 40% to 100% EtOAc/Hexanes) to provide (2S, 3.16 (t, 1H), 2.43 (m, 2H), 1.74 (m. 1H), 1.38 (s, 9H). 4S)-1-tert-butyl 2,4-dimethyl pyrrolidine-1,2,4- 50 tricarboxylate (9.56 g., 94%). (2S,4S)-1-(tert-butoxycarbonyl)-4-(methoxymethyl) pyrrolidine-2-carboxylic acid (3S,5S)-1-(tert-butoxycarbonyl)-5-(methoxycarbo nyl)pyrrolidine-3-carboxylic acid To a solution of (2S,4S)-1-tert-butyl 2-methyl 4-(meth 55 oxymethyl)pyrrolidine-1,2-dicarboxylate (2.11g, 7.7 mmol) To a solution of (2S,4S)-1-tert-butyl 2,4-dimethylpyrroli in a mixture of THF (38 mL) and MeOH (15 mL) was added dine-1,2,4-tricarboxylate (9.56 g., 33.3 mmol) in THF (70 LiOH (2.5 M aqueous, 15 mL, 38.6 mmol). The resulting mL) at 0°C. (external temperature, ice bath) was added solution was stirred at room temperature for 2 h, and acidified NaOH (1N aqueous, 33 mL, 33.3 mmol) dropwise over 15 60 with aqueous HC1 (1N). The desired product was extracted min. The solution was stirred at 0°C. for 5 h before acidifi with CHCl (4x). The combined organic layers were dried cation with HC1 (1N). The solution was extracted with EtOAc over NaSO and concentrated to provide (2S,4S)-1-(tert (3x). The combined organic layers were dried over NaSO butoxycarbonyl)-4-(methoxymethyl)pyrrolidine-2-carboxy and concentrated. The crude oil was purified by silica gel lic acid (2.0 g, 99%). H-NMR: 400 MHz, (CDC1) 8: (mix chromatography (2% to 5% to 10% MeOH/CH2Cl2) to pro 65 ture of rotamers, major reported) 4.33 (t, 1H), 3.65 (m. 1H), vide (3S,5S)-1-(tert-butoxycarbonyl)-5-(methoxycarbonyl) 3.35 (m, 2H), 3.32 (s.3H), 3.16 (t, 1H), 2.45 (m, 2H), 2.12 (m, pyrrolidine-3-carboxylic acid (6.38 g. 70%). 1H), 1.46 (s, 9H).

US 8,940,718 B2 75 76 ethyl)pyrrolidine-1-carboxylate (649 mg 1.25 mmol), bis(pi ((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)pyrroli nacolato)diboron (635 mg, 2.5 mmol), potassium acetate din-2-yl)-1H-imidazol-5-yl)-5H-naphthoc.gchromeno8, (368 mg, 3.7 mmol), tris(dibenzylideneacetone)palladium 9-dimidazol-2-yl)-4-(methoxymethyl)pyrrolidine-1- (46 mg, 0.05 mmol) and 2-dicyclohexylphosphino-2',4',6'- carboxylate (180 mg, 67%). LCMS-ESI: calculated for tri-1-propyl-1,1'-biphenyl (60 mg, 0.12 mmol) in 1,4-dioxane CHN, O, 777.91; observed M+1": 778.84. (7 mL) was heated to 90° C. for 3 hours, cooled to room temperature and diluted with ethyl acetate. The organics were washed with water and brine, dried (Na2SO), and concen methyl 2-2-9-2-(1-2-(methoxycarbonyl) trated. The crude residue was purified by flash chromatogra 10 aminol-3-methylbutanoylpyrrolidin-2-yl)-1H-imi phy to yield (2S,4S)-tert-butyl 4-(methoxymethyl)-2-(9-(4.4, dazol-5-yl)-1,11-dihydroisochromeno4".3':6.7naph 5.5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5H-naphthoc.g tho1,2-dimidazol-2-yl)-4-(methoxymethyl) chromeno8,9-dimidazol-2-yl)pyrrolidine-1-carboxylate pyrrolidin-1-yl)-2-oxo-1-phenylethylcarbamate (467 mg, 61%) LCMS-ESI: calculated for CHBNO: 15 611.54; observed M+1": 612.96. A solution of (2S,4S)-tert-butyl 2-(9-(2-((S)-1-((S)-2- (methoxycarbonylamino)-3-methylbutanoyl)pyrrolidin-2- (2S,4S)-tert-butyl 2-(9-(2-((S)-1-((S)-2-(methoxycar yl)-1H-imidazol-5-yl)-5H-naphthoc.gchromeno8.9-d bonylamino)-3-methylbutanoyl)pyrrolidin-2-yl)-1H imidazol-2-yl)-4-(methoxymethyl)pyrrolidine-1- imidazol-5-yl)-5H-naphthoc.gchromeno8,9-d carboxylate (196 mg, 0.25 mmol), ethanol (3 mL) and imidazol-2-yl)-4-(methoxymethyl)pyrrolidine-1- concentrated HCl (1 mL) was heated to 60°C. for 1 hour. The carboxylate reaction was concentrated and the crude material dissolved in DCM (6 mL). This solution was concentrated and to this material was added a solution of (R)-2-(methoxycarbony To a solution of (2S,4S)-tert-butyl 4-(methoxymethyl)-2- 25 lamino)-2-phenylacetic acid (69 mg 0.33 mmol) and COMU (9-(4.4.5.5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5H-naph (124 mg. 029 mmol) in DMF (4 mL). To the resulting solution thoc.gchromeno8,9-dimidazol-2-yl)pyrrolidine-1-car was added diisopropylethylamine (130 uL, 0.76 mmol). After boxylate (467 mg, 0.76 mmol), methyl (S)-1-(S)-2-(5- stirring for 2 hours at room temperature, the reaction was bromo-1H-imidazol-2-yl)pyrrolidin-1-yl)-3-methyl-1- 30 diluted with ethyl acetate, washed with water and brine, dried oxobutan-2-ylcarbamate (342 mg, 0.92 mmol), tetrakis (NaSO), concentrated and purified by preparative reverse (triphenylphosphine)palladium(0) (44 mg., 0.04 mmol) and phase HPLC (Gemini, 15 to 45% ACN/HO+0.1%TFA). The dichloro1,1'-bis(diphenylphosphino)ferrocenepalladium product fractions were lyophilized to give methyl 2-2-9- (II) (56 mg, 0.07 mmol) in a mixture of 1,2-dimethoxyethane 2-(1-2-[(methoxycarbonyl)aminol-3- (11.0 mL) and dimethylformamide (1.9 mL) was added a 35 methylbutanoylpyrrolidin-2-yl)-1H-imidazol-5-yl)-1,11 solution of potassium carbonate (2M in water, 1.15 mL, 2.29 dihydroisochromeno4".3':6.7naphtho1,2-diimidazol-2- mmol). The resulting mixture was degassed and then heated yl)-4-(methoxymethyl)pyrrolidin-1-yl)-2-oxo-1- to 85°C. under argon for 18 hours. After cooling to room phenylethylcarbamate (84 mg. 39%). LCMS-ESI: temperature, the reaction was diluted with ethyl acetate. The 40 calculated for CashsNOs: 868.98; observed M-- 1: organics were washed with water and brine, dried (NaSO), 870.11 and concentrated. The crude residue was purified by flash chromatography to yield (2S,4S)-tert-butyl 2-(9-(2-((S)-1- Example LS

1. HCl, EtOH, 60° C.

(2S4S)-tert-butyl 2-(9-(2-((S)-1-(S)-2-(methoxycarbonylamino)-3- methylbutanoyl)pyrrollidin-2-yl)-1H-imidazol-5-yl)-5H-naphtho c.gchromeno8,9-dimidazol-2-yl)-4- (methoxymethyl)pyrrollidine-1-carboxylate

US 8,940,718 B2 83 -continued

methyl (2S)-3-methyl-1-oxo-1-(2S)-2-9-(44.5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 3,7-dihydroisochromeno34:5,6naphtho1,2-dimidazol-2-ylpyrrollidin-1-yl)butan-2-yl)carbamate

O Pol(PPh3)4, PdCl2(dppf), y B r se- K2CO3, DME, DMF, 859 C.

O 1) HCI, EtOH, 60° C. Ho N 2) Moc-Val-OH, HATU, DIPEA, DMF

tert-butyl (2R)-2-5-(2-(2S)-1-N-methoxycarbonyl)-L-valylpyrrollidin-2-yl)-3,7- dihydroisochromeno34:5,6naphtho1,2-dimidazol-9-yl)-1H-imidazol-2-ylpyrrollidine-1-carboxylate

methyl (2S)-1-(2R)-2-(5-2-(2S)-1-(2S)-2-(methoxycarbonyl)aminol-3-methylbutanoylpyrrollidin-2-yl)- 3,7-dihydroisochromeno3.4:5,6naphtho1,2-dimidazol-9-yl)-1H-imidazol-2-yl)pyrrollidine-1-yl)- 3-methyl-1-oxobutan-2-yl)carbamate

5-(2-bromo-5-chlorobenzyloxy)-3,4-dihydronaph 8-chloro-2,3,4,6-tetrahydro-1H-dibenzoc.h thalen-1 (2H)-one chromen-1-one To a stirred solution of 5-hydroxy-1-tetralone (2.0 g, 12.3 55 To a flask containing palladium(II) pivalate (68 mg, 0.22 mmol) and 1-bromo-2-(bromomethyl)-4-chlorobenzene (3.6 mmol), tri(4-fluorophenyl)phosphine (70 mg, 0.22 mmol), g, 12.7 mmol) in dimethylformamide (125 mL) was added pivalic acid (135 mg, 1.3 mmol) and potassium carbonate potassium carbonate (3.5g, 25.1 mmol). The reaction was (1.83 g, 13.1 mmol) was added a solution of 5-(2-bromo-5- stirred under argon for 1 hour then diluted with ethyl acetate 60 chlorobenzyloxy)-3,4-dihydronaphthalen-1 (2H)-one (1.61 (1 L). The organics were washed three times with water and g, 4.4 mmol) in dimethyacetamide (23 mL). The flask was once with brine. The organic layer was then dried with mag evacuated and backfilled with argon 5 times and then stirred nesium Sulfate, filtered and concentrated. To the resulting oil under argon at 60° C. for 24 hours. The reaction was poured was added methanol (100 mL) and the Suspension was agi directly onto a silica gel column and purified by flash column tated for thirty minutes. 5-(2-bromo-5-chlorobenzyloxy)-3, 65 chromatography (hexanes/DCM) to yield 8-chloro-2,3,4,6- 4-dihydronaphthalen-1 (2H)-one (4.25 g, 94% yield) was iso tetrahydro-Hi-dibenzoc.hchromen-1-one (1.22 g, 97% lated by filtration. yield) as an off-white solid. US 8,940,718 B2 85 86 2-bromo-8-chloro-2,3,4,6-tetrahydro-1H-dibenzoc, tert-butyl (2S)-2-9-(4.4.5.5-tetramethyl-1,3,2-diox hchromen-1-one aborolan-2-yl)-3,7-dihydroisochromeno3',4': 5.6 naphtho1,2-diimidazol-2-ylpyrrolidine-1-carboxy To a mixture of 8-chloro-2,3,4,6-tetrahydro-1H-dibenzo late c.hchromen-1-one (2.58 g., 9.1 mmol) in chloroform (9.1 To a solution of tert-butyl (2S)-2-(9-chloro-3,7-dihy mL) and ethyl acetate (9.1 mL) was added copper(II) bromide droisochromeno3',4': 5.6naphtho1,2-dimidazol-2-yl)pyr (4.65 g, 19.9 mmol). The reaction was heated to 80°C. for 5 rolidine-1-carboxylate (1.4g, 2.9 mmol) in dioxane (20 mL) hours and then cooled to room temperature. The mixture was was added bis(pinacolato) diboron (1.5 g, 5.9 mmol), tris diluted with dichloromethane and washed twice with a 5:1 (dibenzylideneacetone)dipalladium(0) (110 mg, 0.12 mmol), Solution of Saturated aqueous ammonium chloride and aque 10 X-Phos (145 mg, 0.30 mmol) and potassium acetate (870 mg. ous ammonium hydroxide (-28%), and washed once with 8.9 mmol). The mixture was degassed with a stream of argon water. The organic layer was dried with magnesium Sulfate, for ten minutes. The degassed reaction was heated under filtered and concentrated. The crude material was purified by argon to 90° C. for 2.5 hours then cooled to room temperature flash column chromatography (hexanes/DCM) to yield and diluted with ethyl acetate. The organics were washed with 2-bromo-8-chloro-2,3,4,6-tetrahydro-M-dibenzoch 15 water and brine, dried with magnesium sulfate and concen chromen-1-one (2.45 g, 75% yield). trated. The crude material was purified by flash column chro matography (DCM/EtOAc) to yield tert-butyl (2S)-2-9-(4.4, (2S)-1-tert-butyl 2-(8-chloro-1-oxo-2,3,4,6-tetrahy 5.5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,7- dro-1H-dibenzochchromen-2-yl)pyrrolidine-1,2- dihydroisochromeno3',4': 5.6 naphtho1,2-diimidazol-2-yl) dicarboxylate pyrrolidine-1-carboxylate (1.5g, 90% yield). To a solution of 2-bromo-8-chloro-2,3,4,6-tetrahydro-M- methyl (2S)-3-methyl-1-oxo-1-(2S)-2-9-(4.4.5.5- dibenzochchromen-1-one (1.05g, 2.9 mmol) and Boc-Pro tetramethyl-1,3,2-dioxaborolan-2-yl)-3,7-dihydroiso OH (1.75 g, 8.1 mmol) in acetonitrile (9.0 mL) was added diisopropylethylamine (1.5 mL, 8.7 mmol). The solution was chromeno3',4': 5.6 naphtho1,2-diimidazol-2-yl) stirred under argon at 50° C. for two hours. Extra Boc-Pro 25 pyrrolidin-1-yl)butan-2-yl)carbamate OH (620 mg, 2.9 mmol) and diisopropylethylamine (0.5 mL, 2.9 mmol) were added and the reaction was stirred at 50° C. A solution of tert-butyl (2S)-2-9-(4.4.5.5-tetramethyl-1,3, for 16 hours. The reaction was cooled to room temperature 2-dioxaborolan-2-yl)-3,7-dihydroisochromeno3',4': 5.6 and diluted with ethyl acetate. The organics were washed with naphtho1,2-diimidazol-2-ylpyrrolidine-1-carboxylate water and brine, dried with magnesium Sulfate and concen 30 (0.98g, 1.7 mmol), concentrated HCl (2 mL) and ethanol (20 trated. The crude material was purified by flash column chro mL) was heated to 60° C. for 2 hours. The reaction was matography and the product (2S)-1-tert-butyl 2-(8-chloro-1- concentrated and redissolved in a minimal amount of metha oxo-2,3,4,6-tetrahydro-M-dibenzoc.hchromen-2-yl) nol. An equal Volume of dichloromethane was added and the pyrrolidine-1,2-dicarboxylate was isolated as a mixture of Solution was again concentrated. Dichloromethane was diastereomers (0.99 g, 69% yield). 35 added to the resulting residue and concentrated off two more times. The resulting crude material was dissolved in dimeth tert-butyl (2S)-2-(9-chloro-3,4,5,7-tetrahydroisoch ylformamide (17 mL). To this solution was added (S)-2- romeno3',4': 5.6naphtho1,2-dimidazol-2-yl)pyr (methoxycarbonylamino)-3-methylbutanoic acid (455 mg. rolidine-1-carboxylate 2.6 mmol), HATU (955 mg, 2.5 mmol) and diisopropylethy 40 lamine (3 mL, 17 mmol). The reaction was stirred at room To a solution of (2S)-1-tert-butyl 2-(8-chloro-1-oxo-2,3,4, temperature for one hour then diluted with ethyl acetate. The 6-tetrahydro-H-1-dibenzoc.hchromen-2-yl)pyrrolidine-1, organics were washed with water (x2) and brine, dried with 2-dicarboxylate (2.2g, 4.4 mmol) in toluene (40 mL) was magnesium sulfate and concentrated. The resulting residue added ammonium acetate (7 g., 91 mmol). The reaction mix was purified by flash column chromatography to yield Inter ture was vigorously refluxed for 3 hours, then cooled to room temperature and diluted with ethyl acetate. The organics were 45 mediate methyl (2S)-3-methyl-1-oxo-1-(2S)-2-9-(4.4.5, washed with water and brine, dried with magnesium sulfate 5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,7-dihydroisoch and concentrated. The crude material was purified by flash romeno3',4': 5.6naphtho1,2-dimidazol-2-ylpyrrolidin-1- column chromatography to yield tert-butyl (2S)-2-(9-chloro yl)butan-2-yl)carbamate (780 mg, 72% yield over 2 steps). 3,4,5,7-tetrahydroisochromeno3',4': 5.6 naphtho1,2-dimi dazol-2-yl)pyrrolidine-1-carboxylate (1.13 g, 54% yield) as 50 tert-butyl (2R)-2-5-(2-(2S)-1-N-(methoxycarbo well as recovered (2S)-1-tert-butyl 2-(8-chloro-1-oxo-2,3,4, nyl)-L-valylpyrrolidin-2-yl)-3,7-dihydroisoch 6-tetrahydro-M-dibenzoc.hchromen-2-yl)pyrrolidine-1,2- romeno3',4': 5.6naphtho1,2-dimidazol-9-yl)-1H dicarboxylate (0.8 g., 36%).). LCMS-ESI": calculated for imidazol-2-ylpyrrolidine-1-carboxylate CHNO. 477.98; observed M+1": 478.54. tert-butyl (2S)-2-(9-chloro-3,7-dihydroisochromeno3',4': 5.6 naphtho 55 A mixture of Pentacyclic Intermediate methyl (2S)-3- 1,2-dimidazol-2-yl)pyrrolidine-1-carboxylate methyl-1-oxo-1-(2S)-2-9-(4.4.5.5-tetramethyl-1,3,2-diox To a solution of Intermediate tert-butyl (2S)-2-(9-chloro aborolan-2-yl)-3,7-dihydroisochromeno3',4':5.6naphtho 3,4,5,7-tetrahydroisochromeno3',4':5.6naphtho1,2-dimi 1,2-dimidazol-2-ylpyrrolidin-1-yl)butan-2-yl)carbamate dazol-2-yl)pyrrolidine-1-carboxylate (1.43 g, 3.0 mmol) in (780 mg, 1.3 mmol), (S)-tert-butyl 2-(5-bromo-1H-imidazol dichloromethane (30 mL) was added manganese(IV) oxide 60 2-yl)pyrrolidine-1-carboxylate (450 mg, 1.4 mmol), tetrakis (15g, 198 mmol). The mixture was stirred for four hours at (triphenylphosphine)palladium(0) (30 mg, 0.03 mmol), room temperature then filtered through Celite. The MnO, was PdCl2(dppf) (60 mg, 0.08 mmol), 2M aqueous potassium thoroughly rinsed with dichloromethane and the total filtrate carbonate (1.9 mL, 3.9 mmol), dimethoxyethane (10 mL) and was concentrated to yield tert-butyl (2S)-2-(9-chloro-3,7-di dimethylformamide (2 mL) was degassed with argon for 15 hydroisochromeno3',4': 5.6 naphtho1,2-diimidazol-2-yl) 65 minutes. The reaction was then heated to 85°C. for 3 hours. pyrrolidine-1-carboxylate (1.37 g., 96% yield). This material Upon completion, the reaction was cooled to room tempera was used without further purification. ture, diluted with ethyl acetate and filtered through Celite. US 8,940,718 B2 87 88 The filtrate was washed with water and brine, dried (MgSO) ing residue and concentrated off two more times. One half of and concentrated. The resulting crude material was purified the crude material (-0.27 mmol) was dissolved in dimethyl by flash column chromatography (EtOAc/MeoH) to yield formamide (2.5 mL). To this solution was added (S)-2-(meth Intermediate tert-butyl (2R)-2-5-(2-(2S)-1-N-(methoxy oxycarbonylamino)-3-methylbutanoic acid (66 mg, 0.38 carbonyl)-L-valylpyrrolidin-2-yl)-3,7-dihydroisochromeno 5 mmol), HATU (140 mg, 0.37 mmol) and diisopropylethy 3',4':5.6naphtho1,2-dimidazol-9-yl)-1H-imidazol-2-yl) lamine (0.48 mL, 2.7 mmol). The reaction was stirred at room pyrrolidine-1-carboxylate (390 mg, 43% yield). temperature for 2 hours, and then diluted with acetonitrile (2 mL) and methanol (2 mL). To this solution was added ten methyl {(2S)-1-(2R)-2-(5-2-(2S)-1-(2S)-2- drops of 5M aqueous NaOH solution and stirring was contin (methoxycarbonyl)amino-3- 10 ued for 30 minutes. The reaction was diluted with ethyl methylbutanoylpyrrolidin-2-yl)-3,7-dihydroisoch acetate and the organic layer was washed with water and romeno3',4':5.6naphtho1,2-dimidazol-9-yl)-1H brine. The combined aqueous washings were extracted three imidazol-2-yl)pyrrolidin-1-yl)-3-methyl-1-oxobutan times with ethyl acetate, and the combined organic layers 2-yl)carbamate were dried (MgSO) and concentrated. The crude material 15 was purified by reverse phase HPLC (Gemini, 15 to 45% A mixture of Intermediate tert-butyl (2R)-2-5-(2-(2S)-1- ACN/HO+0.1% TFA) to yield methyl (2S)-1-(2R)-2-(5- N-(methoxycarbonyl)-L-valylpyrrolidin-2-yl)-3,7-dihy {2-(2S)-1-(2S)-2-[(methoxycarbonyl)aminol-3- droisochromeno3',4':5.6 naphtho1,2-diimidazol-9-yl)-1H methylbutanoylpyrrolidin-2-yl)-3,7-dihydroisochromeno imidazol-2-ylpyrrolidine-1-carboxylate (390 mg, 0.53 3',4': 5.6naphtho1,2-dimidazol-9-yl)-1H-imidazol-2-yl) mmol), concentrated HCl (2 mL) and ethanol (10 mL) was pyrrolidin-1-yl)-3-methyl-1-oxobutan-2-yl)carbamate (140 heated to 60° C. for 2 hours. The reaction was concentrated mg, 67% yield over 2 steps). LCMS-ESI: calculated for and redissolved in a minimal amount of methanol. An equal CHNO: 790.91; observed M+1": 791.71. Volume of dichloromethane was added and the solution was again concentrated. Dichloromethane was added to the result Example MA NO

methyl (1R)-2-(2R)-2-(5-2-(2S)-1-(2S)-2-[(methoxycarbonyl)aminol-3-methylbutanoylpyrrollidin-2-yl)- 3,7-dihydroisochromeno34:5,6naphtho1,2-dimidazol-9-yl)-1H-imidazol-2-yl)pyrrollidin-1-yl)- 2-oxo-1-phenylethyl)carbamate

This compound was made in an analogous manner to 40 methyl (2S)-1-(2R)-2-(5-2-(2S)-1-(2S)-2-[(methoxy carbonyl)aminol-3-methylbutanoylpyrrolidin-2-yl)-3,7-di hydroisochromeno3',4': 5.6naphtho1,2-dimidazol-9-yl)- 1H-imidazol-2-yl)pyrrolidin-1-yl)-3-methyl-1-oxobutan-2- y1}carbamate, substituting (R)-2-(methoxycarbonylamino)- 45 2-phenylacetic acid for (S)-2-(methoxycarbonylamino)-3- methylbutanoic acid and substituting COMU for HATU in the final amide coupling step. LCMS-ESI: calculated for C.H.N.O. 824.92; observed M+1": 825.72. Example MB

NO 2-y O O Y- Pd(PPh3)4, NH O PdCl2(dppf), Her )- y K2CO3, N U. N N DME, DMF, 850 C.

methyl (S)-1-(S)-2-(5-bromo-1H tert-butyl (2S)-2-9-(44.5,5-tetramethyl-1,3,2-dioxaborolan imidazol-2-yl)pyrrollidin-1-yl)-3- 2-yl)-3,7-dihydroisochromeno34:5,6naphtho1,2- methyl-1-oxobutan-2-ylcarbamate dimidazol-2-ylpyrrollidine-1-carboxylate US 8,940,718 B2 90 -continued

tert-butyl (2S)-2-9-(2-(2R)-1-N-(methoxycarbonyl)-L- valylpyrrollidin-2-yl)-1H-imidazol-5-yl)-3,7- dihydroisochromeno34:5,6naphtho1,2-dimidazol-2- ylpyrrollidine-1-carboxylate in-QO 1) HCI, EtOH, 60° C. -e- 2) Moc-Val-OH, HATU, Y- O DIPEA, DMF HO

(R)-2-(methoxycarbonylamino)- 2-phenylacetic acid

O O

- N

methyl (2S)-1-(2R)-2-(5-2-(2S)-1-(2R)-2-[(methoxycarbonyl)amino)-2-phenylacetylpyrrollidin-2-yl)-3,7- dihydroisochromeno34:5,6naphtho1,2-dimidazol-9-yl)-1H-imidazol-2-yl)pyrrollidin-1-yl)-3- methyl-1-oxobutan-2-yl)carbamate

tert-butyl (2S)-2-9-(2-(2R)-1-N-(methoxycarbo methyl (2S)-1-(2R)-2-(5-2-(2S)-1-(2R)-2- nyl)-L-valylpyrrolidin-2-yl)-1H-imidazol-5-yl)-3,7- 40 (methoxycarbonyl)amino-2- dihydroisochromeno3',4': 5.6naphtho1,2-dimida phenylacetylpyrrolidin-2-yl)-3,7-dihydroisoch zol-2-ylpyrrolidine-1-carboxylate romeno3',4': 5.6naphtho1,2-dimidazol-9-yl)-1H imidazol-2-yl)pyrrolidin-1-yl)-3-methyl-1-oxobutan 45 2-yl)carbamate A mixture of tert-butyl (2S)-2-9-(4.4.5.5-tetramethyl-1,3, 2-dioxaborolan-2-yl)-3,7-dihydroisochromeno3',4': 5.6 A mixture of tert-butyl (2S)-2-9-(2-(2R)-1-N-(meth naphtho1,2-dimidazol-2-ylpyrrolidine-1-carboxylate (450 oxycarbonyl)-L-valylpyrrolidin-2-yl)-1H-imidazol-5-yl)- mg, 0.79 mmol), methyl (S)-1-((S)-2-(5-bromo-1H-imida 3,7-dihydroisochromeno3',4': 5.6naphtho1,2-dimidazol Zol-2-yl)pyrrolidin-1-yl)-3-methyl-1-oxobutan-2-ylcarbam 50 2-ylpyrrolidine-1-carboxylate (270 mg, 0.37 mmol), concentrated HCl (1.5 mL) and ethanol (8 mL) was heated to ate (325 mg. 0.87 mmol), tetrakis(triphenylphosphine)palla 60° C. for 1 hour. The reaction was concentrated and redis dium(0) (30 mg, 0.02 mmol), PdCl(dppf) (35 mg, 0.05 Solved in a minimal amount of methanol. An equal Volume of mmol), 2M aqueous potassium carbonate (1.2 mL, 2.4 dichloromethane was added and the Solution was again con mmol), dimethoxyethane (6.8 mL) and dimethylformamide 55 centrated. Dichloromethane was added to the resulting resi (1.2 mL) was degassed with argon for 15 minutes. The reac due and concentrated off two more times. The crude material tion was then heated to 85°C. for 2.5 hours. Upon comple was dissolved in 5:1 dichloromethane/dimethylformamide tion, the reaction was cooled to room temperature, diluted (3.8 mL). To this solution was added (R)-2-(methoxycarbo with ethyl acetate and filtered through Celite. The filtrate was nylamino)-2-phenylacetic acid (96 mg, 0.46 mmol), COMU 60 (190 mg, 0.44 mmol) and diisopropylethylamine (0.20 mL. washed with water and brine, dried (MgSO) and concen 1.1 mmol). The reaction was stirred at 0°C. for 30 minutes trated. The resulting crude material was purified by flash then warmed to room temperature. Upon completion, the column chromatography (EtOAc/MeOH) to yield tert-butyl reaction was diluted with acetonitrile (2 mL) and methanol (2 (2S)-2-9-(2-(2R)-1-N-(methoxycarbonyl)-L-valylpyrro mL). To this solution was added ten drops of 5M aqueous lidin-2-yl)-1H-imidazol-5-yl)-3,7-dihydroisochromeno3', 65 NaOH solution and stirring was continued for 30 minutes. 4': 5.6naphtho1,2-dimidazol-2-ylpyrrolidine-1-carboxy The reaction was diluted with ethyl acetate and the organic late (270 mg, 46% yield). layer was washed with water and brine. The combined aque

US 8,940,718 B2 135 136 min, then heated to 100° C. for 16 h. The solution was cooled to rt, diluted with EtOAc, washed with saturated aqueous (2S,4S)-1-tert-butyl 2-(3-chloro-8-oxo-8,9,10,11 NaHCO, brine, dried with MgSO, and concentrated. Puri tetrahydro-5H-dibenzoc.gchromen-9-yl) 4-meth fied by silica gel chromatography (0% to 30% MeOH/EtOAc) ylpyrrolidine-1,2-dicarboxylate to afford (2S,4S)-tert-butyl 4-methyl-2-(9-(4.4.5.5-tetram ethyl-1,3,2-dioxaborolan-2-yl)-1,11-dihydroisochromeno To a solution of 9-bromo-3-chloro-10,11-dihydro-5H 4',3':6.7naphtho1,2-dimidazol-2-yl)pyrrolidine-1-car dibenzoc.gchromen-8(9H)-one (1.32 g, 3.63 mmol) in boxylate (821 mg, quant). MeCN (40 mL) was added (2S,4S)-1-(tert-butoxycarbonyl)- 4-methylpyrrolidine-2-carboxylic acid (1.0 g, 4.36 mmol) 10 and DIPEA (0.7 mL, 3.99 mmol). After stirring for 18 h, the (2S,4S)-tert-butyl 2-(9-(2-08)-1-08)-2-(methoxycar solution was diluted with EtOAc and washed successively bonylamino)-3-methylbutanoyl)pyrrolidin-2-yl)-1H with saturated aqueous NaHCO and brine. The organics imidazol-5-yl)-5H-naphthoc.gchromeno8.9-d were dried over MgSO filtered and concentrated under imidazol-2-yl)-4-(methyl)pyrrolidine-1-carboxylate reduced pressure. The crude residue was purified by silica 15 column chromatography (10% to 40% EtOAc/hexanes) to To a solution of (2S,4S)-tert-butyl 4-methyl-2-(9-(4.4.5.5- afford (2S,4S)-1-tert-butyl 2-(3-chloro-8-oxo-8,9,10,11-tet tetramethyl-1,3,2-dioxaborolan-2-yl)-1,11-dihydroisoch rahydro-5H-dibenzoc.gchromen-9-yl) 4-methylpyrroli romeno4".3':6.7naphtho1,2-dimidazol-2-yl)pyrrolidine dine-1,2-dicarboxylate (1.31 g, 70%). 1-carboxylate (821 mg, 1.41 mmol), methyl (S)-1-((S)-2-(5- bromo-1H-imidazol-2-yl)pyrrolidin-1-yl)-3-methyl-1- (2S,4S)-tert-butyl 2-(9-chloro-4,5-dihydro-5H-naph oxobutan-2-ylcarbamate (1.05 g, 2.82 mmol), tetrakis thoc.gchromeno8,9-dimidazol-2-yl)-4-methylpyr (triphenylphosphine)palladium(0) (162 mg, 0.14 mmol) and rolidine-1-carboxylate dichloro1,1'-bis(diphenylphosphino)ferrocenepalladium (2S,4S)-1-tert-butyl 2-(3-chloro-8-oxo-8.9.10.11-tetrahy 25 (II) (102 mg, 0.14 mmol) in DME (15 mL) was added a dro-5H-dibenzoc.gchromen-9-yl) 4-methylpyrrolidine-1, solution of potassium carbonate (2M in water, 2.32 mL, 4.65 2-dicarboxylate (1.31 g, 2.56 mmol) was added xylenes (25 mmol). The resulting mixture was degassed and then heated mL) and ammonium acetate (3.95 g, 51.2 mmol) and the to 85°C. for 18 hours. After cooling to room temperature, the solution was heated to 136° C. and stirred overnight. The reaction was diluted with ethyl acetate. The organics were following morning, the Solution was cooled to rt and was 30 washed with saturated sodium bicarbonate and brine, dried diluted with EtOAc and washed successively with water, over MgSO and concentrated. The crude residue was puri saturated aqueous NaHCO and brine. The organics were fied by flash chromatography to yield (2S,4S)-tert-butyl 2-(9- dried over MgSO, filtered and concentrated under reduced (2-((S)-1-((S)-2-(methoxycarbonylamino)-3-methylbu pressure. The crude residue was purified by silica column tanoyl)pyrrolidin-2-yl)-1H-imidazol-5-yl)-5H-naphthoc.g chromatography (60% to 100% EtOAc/hexanes) to afford 35 chromeno8,9-dimidazol-2-yl)-4-methylpyrrolidine-1- (2S,4S)-tert-butyl 2-(9-chloro-4,5-dihydro-5H-naphthoc.g carboxylate (386 mg, 37%). chromeno8,9-dimidazol-2-yl)-4-methylpyrrolidine-1-car boxylate (711 mg, 56%). Methyl 2-2-9-2-(1-2-(methoxycarbonyl) (2S,4S)-tert-butyl 2-(9-chloro-5H-naphthoc.g 40 aminol-3-methylbutanoylpyrrolidin-2-yl)-1H-imi chromeno8,9-dimidazol-2-yl)-4-methylpyrrolidine dazol-5-yl)-1,11-dihydroisochromeno4".3':6.7naph 1-carboxylate tho1,2-dimidazol-2-yl)-4-(methyl)pyrrolidin-1-yl)- 2-oxo-1-phenylethylcarbamate To a solution of (2S,4S)-tert-butyl 2-(9-chloro-4,5-dihy dro-5H-naphthoc.gchromeno8,9-dimidazol-2-yl)-4-(me 45 A solution of (2S,4S)-tert-butyl 2-(9-(2-((S)-1-((S)-2- thyl)pyrrolidine-1-carboxylate (93.5 mg, 1.9 mmol) in (methoxycarbonylamino)-3-methylbutanoyl)pyrrolidin-2- CHCl (20 mL) was added MnO, (8.25 g, 95 mmol). The yl)-1H-imidazol-5-yl)-5H-naphthoc.gchromeno8.9-d reaction mixture was stirred for 3 h, and then filtered over celite. The filter cake was washed with copious CHCl and imidazol-2-yl)-4-methylpyrrolidine-1-carboxylate (386 mg, MeOH, and the filtrate was concentrated under reduced pres 50 0.52 mmol), CHCl (8 mL), MeOH (2 mL) and HCl (4M in sure. The crude residue was purified by silica column chro Dioxane, 2 mL) and was stirred overnight. The reaction was matography (0% to 10% MeOH/EtOAc) to afford (2S,4S)- concentrated and the crude material dissolved in DMF (8 tert-butyl 2-(9-chloro-5H-naphthoc.gchromeno8.9-d mL). This solution was concentrated and to this material was imidazol-2-yl)-4-methylpyrrolidine-1-carboxylate (692 mg, added a solution of (R)-2-(methoxycarbonylamino)-2-phe 74%). 55 nylacetic acid (108 mg, 0.52 mmol) and COMU (248 mg, 0.52 mmol). To the resulting solution was added diisopropy (2S,4S)-tert-butyl 4-methyl-2-(9-(4,4,5,5-tetram lethylamine (0.45 mL, 2.6 mmol). After stirring for 2 hours at ethyl-1,3,2-dioxaborolan-2-yl)-1,11-dihydroisoch room temperature, the reaction was diluted with 10% MeOH/ romeno4".3':6.7naphtho1,2-dimidazol-2-yl)pyrro EtOAc, washed with saturated NaHCO, water and brine, lidine-1-carboxylate 60 dried (NaSO), concentrated and purified by HPLC to give methyl 2-2-9-2-(1-2-(methoxycarbonyl)aminol-3- (2S,4S)-tert-butyl 2-(9-chloro-5H-naphthoc.gchromeno methylbutanoylpyrrolidin-2-yl)-1H-imidazol-5-yl)-1,11 8,9-dimidazol-2-yl)-4-methylpyrrolidine-1-carboxylate dihydroisochromeno4".3':6.7naphtho1,2-diimidazol-2- (692 mg, 1.41 mmol) in dioxane (15 mL) was added bis yl)-4-methylpyrrolidin-1-yl)-2-oxo-1- (pinacolato) diboron (1.07 g. 4.23 mmol), KOAc (415 mg. 65 phenylethylcarbamate (27 mg, 6%). LCMS-ESI". 4.23 mmol), X-Phos (52 mg, 0.11 mmol), and Pd dba (26 calculated for C7H5NO7: 838.38; observed M--1": mg, 0.03 mmol). The solution was degassed with N for 10 840.12

US 8,940,718 B2 174 -continued (3x). Palladium on carbon (apprx. 750 mg, 10% w/w, dry) Na2SO3 was added and the reaction was evacuated of gas and charged IIICH --> with hydrogen gas (3x). The reaction was allowed to stir I N * Dioxane, H2O Ns' N under atmospheric hydrogen for 16 hours. The mixture was \ V filtered through a plug of celite and the filtrate was concen NH Boc trated in vacuo. Diethyl ether was added to the oil and a precipitate formed. The mixture was filtered to yield (2S,5S)- I ethyl 5-methylpyrrolidine-2-carboxylate, as a white solid (2S,5S)-tert-butyl 2 (10.6 g. 67.4 mmol. 86.7% over three steps). H NMR (400 (4,5-diiodo-1H MHz, cdcl.) & 4.48 (dd. 1H), 4.27 (q, 2H), 3.92-3.80 (m. 1H), imidazol-2-yl)- 10 5-methylpyrrollidine 2.52-2.36 (m. 1H), 2.32-2.13 (m, 2H), 1.75-1.60 (m, 1H), 1-carboxylate 1.51 (d. 3H), 1.30 (t, 3H). 1.HC Dioxane (2S,5S)-1-tert-butyl 2-ethyl II ICH -- N s' 2. L-Valine MOC 5-methylpyrrolidine-1,2-dicarboxylate y N HATU, DIEA 15 DCM To a solution of (2S,5S)-ethyl 5-methylpyrrolidine-2-car \ NH Boc boxylate (7.0 g, 44.5 mmol) in dichloromethane (250 mL), I ditertbutylanhydride (10.7 g. 49.0 mmol), diisopropylethy (2S,5S)-tert-butyl 2 lamine (17.1 mL. 98.0 mmol) dropwise over 10 minutes, and (5-iodo-1H dimethyl amino pyridine (0.27 g. 2.23 mmol) were added imidazol-2-yl)- successively. Effervescence was observed and the mixture 5-methylpyrrollidine was allowed to stir for 16 hours at room temperature. The 1-carboxylate O) w reaction was washed with HCl (250 mL, of 1N). The organic N f layer was then dried with sodium sulfate. The crude material 25 was purified by column chromatography (5%-25% EtOAc/ hexanes) to yield (2S,5S)-1-tert-butyl 2-ethyl 5-methylpyrro \ YY S\ lidine-1,2-dicarboxylate as an oil (6.46g, 25.1 mmol. 56%). I NH O s O LCMS-ESI: calc'd for CHNO: 257.16 (M"). Found: 258.70 (M+H"). 30 (2S,5S)-1-(tert-butoxycarbonyl)-5-methylpyrroli dine-2-carboxylic acid 5-methylpyrrollidin-1-yl)-2- To a solution of (2S,5S)-1-tert-butyl 2-ethyl 5-methylpyr (1-methoxyethenyl)amino rolidine-1,2-dicarboxylate (6.46g, 25.1 mmol) in ethanol (20 3-methylbutan-1-one 35 mL) was added lithium hydroxide monohydrate (2.11g, 50.2 mmol) and deionized water (12 mL). The mixture was (S)-ethyl allowed to stir for 16 hours then partitioned between ethylac 2-(tert-butoxycarbonylamino)-5-oxohexanoate etate and a 1:1 mixture of saturated brine and 1N HC1. The aqueous layer was extracted an additional time with ethyl A solution of ethyl N-Boc (S)-pyroglutamate (20.0 g, 77.7 40 acetate. The organic layers were combined, dried with mmol) was in anhydrous THF (150 mL) in a two neck round sodium sulfate and the solvent was removed in vacuo to yield bottom under argon was cooled to -40°C. Methyl-magne (2S,55)-1-(tert-butoxycarbonyl)-5-methylpyrrolidine-2-car sium bromide solution (3.0M in Ether, 28.5 mL, 85.5 mmol) boxylic acid as a white solid (quant.) and was used directly in was added to the reaction mixture dropwise over 30 minutes. the following step. The reaction was stirred for 4 hrs at -40°C. then for 1 hr at 0° 45 C. The reaction was partitioned between ethyl acetate and (2S,5S)-tert-butyl 2-(hydroxymethyl)-5-methylpyr saturated ammonium chloride solution and acidified with 1 N rolidine-1-carboxylate HC1. The aqueous layer was extracted two more times with ethylacetate. The organic layers were combined and dried with sodium sulfate. The crude material was purified by col To a solution of (2S,5S)-1-(tert-butoxycarbonyl)-5-meth umn chromatography (20%-40%. EtOAc/hexanes) to yield 50 ylpyrrolidine-2-carboxylic acid (5.91 g, 25.8 mmol) in tet (S)-ethyl 2-(tert-butoxycarbonylamino)-5-oxohexanoate as a rahydrofuran at 0°C., was added borane in dimethylsulfide Viscous oil and was used directly in the following step. (1.0 M, 3.4 mL, 34 mmol) dropwise. The reaction was stirred for 4 hours at 0°C. then 18 hours at room temperature. The (S)-ethyl mixture was then cooled to 0°C. and methanol (70 mL) was 5-methyl-3,4-dihydro-2H-pyrrole-2-carboxylate added dropwise. The reaction was warmed to room tempera 55 ture and the solvents were removed in vacuo. The residue was (S)-ethyl 2-(tert-butoxycarbonylamino)-5-oxohexanoate taken up in dichloromethane (200 mL) and extracted with in a 1 L flask was treated with a trifluoro acetic acid/dichlo saturated sodium bicarbonate. The organic layer was dried romethane solution (1:1 mixture, 100 mL). Effervescence with sodium sulfate and the solvent was removed in vacuo to was observed and the mixture was allowed to stir for 4 hours yield (2S,55)-tert-butyl 2-(hydroxymethyl)-5-methylpyrroli at room temperature. After which time the volatiles were 60 removed in vacuo to yield (S)-ethyl 5-methyl-3,4-dihydro dine-1-carboxylate as a clear oil (5.15g, 23.9 mmol. 93%) 2H-pyrrole-2-carboxylate as an oil, and used directly in the and was used directly in the following step. following step. (2S,5S)-tert-butyl (2S,5S)-ethyl 5-methylpyrrolidine-2-carboxylate 2-formyl-5-methylpyrrolidine-1-carboxylate 65 The crude imine in a 1L flask was dissolved with ethanol To a solution of (2S,55)-tert-butyl 2-(hydroxymethyl)-5- (400 mL) was evacuated and charged with argon three times methylpyrrolidine-1-carboxylate (5.15 g, 23.9 mmol) in US 8,940,718 B2 175 176 dichloromethane, was added TEMPO (0.075 g, 0.48 mmol), hexanes to yield (2S,5S)-tert-butyl 2-(4,5-diiodo-1H-imida sodium bromide (0.246g, 2.39 mmol) and sodium bicarbon Zol-2-yl)-5-methylpyrrolidine-1-carboxylate as a white solid ate (0.442 g, 5.26 mmol). Sodium hypochlorite (2.67 g., 35.9 (4.28 g, 8.50 mmol, 62%). LCMS-ESI": calc'd for mmol) of a 6% solution was added and the biphasic mixture C.H.I.N.O.: 502.96 (M). Found: 503.94 (M+H"). was vigorously stirred for 2 hours at room temperature. The (2S,5S)-tert-butyl 2-(5-iodo-1H-imidazol-2-yl)-5- reaction mixture was extracted two times with dichlo methylpyrrolidine-1-carboxylate romethane (2x100 mL). The organic layers were combined and washed with saturated sodium thiosulfate solution, dried To a solution of (2S,5S)-tert-butyl 2-(4,5-diiodo-1H-imi with sodium sulfate and the solvent was removed in vacuo to dazol-2-yl)-5-methylpyrrolidine-1-carboxylate (4.28 g, 8.50 yield (2S,5S)-tert-butyl 2-formyl-5-methylpyrrolidine-1-car 10 mmol) in ethanol (75 mL) and water (75 mL), sodium thio boxylate (3.9 g, 18.29 mmol. 77%) as a slight colored oil and sulfate (10.72 g, 85.1 mmol) was added and the reaction was used directly in the following step. mixture was stirred vigorously for 1 hour at 100° C., 16 hours at 90° C., and 5 hours at 100° C. The reaction mixture was (2S,5S)-tert-butyl 2-(1H-imidazol-2-yl)-5-meth partitioned between ethyl acetate and water. The aqueous ylpyrrolidine-1-carboxylate 15 layer was washed additionally with ethyl acetate and the organic layers were combined. The organic layer was dried To a solution of (2S,55)-tert-butyl 2-formyl-5-methylpyr with sodium sulfate, concentrated and the crude material was rolidine-1-carboxylate (3.9 g, 18.30 mmol) in MeOH (15 mL) purified by column chromatography to yield (2S,5S)-tert and ammonium hydroxide (15 mL. 99.9%), glyoxal (11.7 butyl 2-(5-iodo-1H-imidazol-2-yl)-5-methylpyrrolidine-1- mL, 40% w/v in water, 102.40 mmol) was added dropwise. carboxylate as a white solid (2.34 g. 6.20 mmol. 73%). "H The biphasic mixture turned orange and turbid. The reaction NMR (400 MHz, cdcl.) & 7.04 (s, 1H), 4.89 (dd. 1H), 3.92 (m, was stirred vigorously overnight at room temperature. The 1H), 2.91 (s, 1H), 2.18-2.06 (m, 2H), 1.78 (m, 1H), 1.52 (m, solvent was removed in vacuo. The crude mixture was redis 1H), 1.48 (s, 9H), 1.13 (d. 3H). solved in ethyl acetate and washed with water. The aqueous layer was washed an additional time with ethyl acetate. The (2S)-1-(2S,5S)-2-(5-iodo-1H-imidazol-2-yl)-5-me organic layers were combined and washed with brine, dried 25 thylpyrrolidin-1-yl)-2-(1-methoxyethenyl)amino-3- with sodium sulfate and the solvent was removed in vacuo. methylbutan-1-one The crude material was purified by column chromatography 85% to 100% ethyl acetate in hexanes to yield (2S,5S)-tert A round bottom flask was charged with (2S,5S)-tert-butyl butyl 2-(1H-imidazol-2-yl)-5-methylpyrrolidine-1-carboxy 2-(5-iodo-1H-imidazol-2-yl)-5-methylpyrrolidine-1-car late as an off white solid (3.47 g. 13.8 mmol, 75%). LCMS 30 boxylate (1.5 g, 3.98 mmol) and treated with an excess of ESI: calc’d for CHNO: 251.16 (M"). Found: 252.20 hydrochloric acid (100 mL of 4.0M in dioxane). The mixture (M+H"). was stirred vigorously for 3 hours in which time a precipitate formed and the solvent was removed in vacuo. To a mixture of (2S,5S)-tert-butyl 2-(4,5-diiodo-1H-imidazol-2-yl)- the crude intermediate, (S)-2-(methoxycarbonylamino)-3- 5-methylpyrrolidine-1-carboxylate 35 methylbutanoic acid (0.836 g. 4.77 mmol), HATU (1.81 g, 4.77 mmol) in dichloromethane (25 mL), diisopropylethy A 500 mL round bottom flask was charged with (2S,5S)- lamine (3.46 mL, 19.9 mmol) was then added dropwise and tert-butyl 2-(1H-imidazol-2-yl)-5-methylpyrrolidine-1-car was stirred over night under nitrogen. The reaction mixture boxylate (3.47 g. 13.8 mmol), iodine (7.7g, 30.4 mmol) and was partitioned ethyl acetate and Saturated Sodium bicarbon sodium carbonate (4.54g, 42.8 mmol). Dioxane (70 mL) and 40 ate. The organic layer was dried with sodium sulfate, the water (45 mL) was added to mixture and the reaction was solvent removed in vacuo. The crude product was purified by stirred vigorously overnight in the dark. The reaction was column chromatography to yield (2S)-1-(2S,5S)-2-(5-iodo then partitioned between ethyl acetate and a 10% aqueous 1H-imidazol-2-yl)-5-methylpyrrolidin-1-yl)-2-(1-meth Solution of sodium thiosulfate and extracted. The aqueous oxyethenyl)aminol-3-methylbutan-1-one as a white solid layer was extracted an additional time with ethyl acetate. The (1.63 g, 3.75 mmol, 94%). LCMS-ESI: calc'd for organic layers were combined, dried with sodium sulfate and 45 CHINO: 434.08 (M). Found: 435.51 (M+H"). the solvent was removed in vacuo. The crude material was filtered through a plug of silica with 25% ethyl acetate in Example NP

Pol(PPh3)4, PdCl2(dppf),

(2S, 4S)-tert-butyl 4-(methoxymethyl)-2-(9- (44.5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 5 H-naphthoc, gchromeno8,9-dimidazol-2-yl) pyrrollidine-1-carboxylate

US 8,940,718 B2 183 184 -continued thylbutanoic acid (6.88 g. 39.3 mmol) and HATU (14.9 g, NHCOMe 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 O LiOH phase was washed successively with 10% HCl, saturated He HO/MeOH aqueous NaHCO and brine, then dried over MgSO, filtered and concentrated under reduced pressure to afford (2S,5S)- EtO) - ethyl 1-((S)-2-(methoxycarbonylamino)-3-methylbutanoyl)- (2S,5S)-ethyl 1-(S)-2- 5-methylpyrrolidine-2-carboxylate. The crude material was (methoxycarbonylamino)-3- 10 methylbutanoyl)-5- carried on without further purification. methylpyrrollidine-2-carboxylate (2S,5S)-1-((S)-2-(Methoxycarbonylamino)-3-meth NHCOMe ylbutanoyl)-5-methylpyrrolidine-2-carboxylic acid 15 (2S,5S)-Ethyl 1-((S)-2-(methoxycarbonylamino)-3-meth N ylbutanoyl)-5-methylpyrrolidine-2-carboxylate (39.3 mmol. assuming complete conversion from the previous transforma tion) was suspended in MeOH (200 mL) and aqueous LiOH HO) k (1.0M, 100 mL, 100 mmol) was added. The reaction mixture (2S,5S)-1-((S)-2- was stirred of n, then concentrated under reduced pressure to (methoxycarbonylamino)-3- remove most of the MeCH. The aqueous solution was washed methylbutanoyl)-5- 2x with DCM before being acidified to pH-1-2 with 10% methylpyrrollidine-2-carboxylic acid HC1. The acidic aqueous phase was then extracted 5x with EtOAc. The combined EtOAc extracts were dried over 25 MgSO filtered and concentrated under reduced pressure to (2S,5S)-Ethyl 1-((S)-2-(methoxycarbonylamino)-3- afford (2S,55)-1-(S)-2-(Methoxycarbonylamino)-3-meth methylbutanoyl)-5-methylpyrrolidine-2-carboxylate ylbutanoyl)-5-methylpyrrolidine-2-carboxylic acid (6.89 g, 56% over 2 steps). (2S,5S)-Ethyl 5-methylpyrrolidine-2-carboxylate-TFA (10.0 g, 39.3 mmol), (S)-2-(methoxycarbonylamino)-3-me Example OF

potassium O vinyltrifluoroborate, O O Pd(OAc), SPhos, O 1. NBS K2CO3 H2O/THF/DMSO -e- -e- propanol (reflux) / ( ) 2. MnO, DCM

3-chloro-10,11-dihydro-5H-dibenzoc.g 3-vinyl-10,11-dihydro-5H-dibenzoc.g.) chromen-8(9H)-one chromen-8(9H)-one

HOC Roc Cs2CO3 -- -e- 2-Me-THF

OMe 3-(2-bromoacetyl)-10,11-dihydro-5H-dibenzoc.g (2S4S)-1-(tert-butoxycarbonyl)- chromen-8(9H)-one 4-(methoxymethyl)pyrrollidine-2- carboxylic acid MeO O pyridinium tribromide, DCMJMeOH Yy O O Hibs NBoc O ( ) ( () ) (2S4S)-1-tert-butyl 2-(2-oxo-2-(8-oxo-8,9,10,11-tetrahydro-5H dibenzoc.gchromen-3-yl)ethyl)-4-(methoxymethyl)pyrrollidine-1,2-dicarboxylate

US 8,940,718 B2 187 -continued

methyl (2S,3S)-1-(2S4S)-2-(5-2-(2S,5S)-1-(2S)-2-[(methoxycarbonyl)aminol-3-methylbutanoyl-5- methylpyrrollidin-2-yl)-1,11-dihydroisochromeno4',3':6.7naphtho1,2-dimidazol-9-yl)-1H imidazol-2-yl)-4-(methoxymethyl)pyrrollidin-1-yl)-3-methyl-1-oxopentan-2-yl)carbamate

3-Vinyl-10,11-dihydro-5H-dibenzoc.gchromen-8 butoxycarbonyl)-4-(methoxymethyl)pyrrolidine-2-carboxy (9H)-one 2O lic acid (5.22 g. 20.1 mmol) were suspended in 2-Me-THF A 3-neck oven-dried 500 mL round-bottom flask was (75 mL) and treated with CsCO (3.27 g. 10.1 mmol). After cooled under Ar, then charged with 3-Chloro-10,11-dihydro stirring 4hat RT, the reaction mixture was diluted with DCM. 5H-dibenzoc.gchromen-8(9H)-one (12.0 g, 42.1 mmol), The organic layer was washed with H2O. The aqueous layer potassium vinyltrifluoroborate (8.47 g. 6.32 mmol), was then back extracted 2x with DCM. The combined organ Pd(OAc) (473 mg, 2.11 mmol), SPhos (1.74g, 4.25 mmol), 25 ics were dried over MgSO, filtered and concentrated under KCO (17.5 g. 126 mmol) and anhydrous propanol (120 reduced pressure. The crude residue was purified by silica mL). The reaction mixture was sparged with Ar for 16 min, column chromatography (10% to 50% EtOAc/DCM) to then heated to reflux for 5.5h. Upon completion, the reaction afford (2S,4S)-1-tert-butyl 2-(2-oxo-2-(8-oxo-8,9,10,11-tet mixture was cooled to RT and concentrated under reduced rahydro-5H-dibenzoc.gchromen-3-yl)ethyl) 4-(meth pressure. The crude residue was suspended in DCM, then 30 oxymethyl)pyrrolidine-1,2-dicarboxylate (7.73 g, 70%). washed with HO and brine. The organic solution was dried over MgSO filtered and concentrated under reduced pres (2S,4S)-2-(2-(9-Bromo-8-oxo-8.9.10, 11-tetrahydro Sure. The resulting residue was further purified via silica plug, 5H-dibenzoc.gchromen-3-yl)-2-oxoethyl) 1-tert eluting with DCM to afford 3-vinyl-10,11-dihydro-5H butyl 4-(methoxymethyl)pyrrolidine-1,2-dicarboxy dibenzoc.gchromen-8(9H)-one (10.2g, 87%). 35 late 3-(2-Bromoacetyl)-10,11-dihydro-5H-dibenzoc.g (2S,4S)-1-tert-Butyl 2-(2-oxo-2-(8-oxo-8,9,10,11-tet chromen-8(9H)-one rahydro-5H-dibenzoc.gchromen-3-yl)ethyl) 4-(meth oxymethyl)pyrrolidine-1,2-dicarboxylate (7.66 g. 13.9 3-Vinyl-10,11-dihydro-5H-dibenzoc.gchromen-8(9H)- 40 mmol) was dissolved in a solution of DCM (100 mL) and one (9.98 g., 36.1 mmol) was dissolved in a stirred solution of MeOH (40 mL), then treated with pyridinium tribromide THF (70 mL), DMSO (70 mL) and HO (35 mL). NBS (6.75 (4.90 g, 15.3 mmol). After stirring at RT for 1.75 h, the g, 37.9 mmol) was added in a single portion and the reaction reaction mixture was diluted with DCM and washed succes mixture was stirred at RT for 33 min. Upon completion, the sively with 10% HCl, saturated aqueous NaHCO and brine. reaction medium was diluted with EtOAc and washed twice 45 The organic phase was dried over MgSO filtered and con with HO and once with brine. The organic phase was dried centrated under reduced pressure and the crude material was over MgSO filtered and concentrated under reduced pres carried on without further purification. Sure. The resulting crude bromohydrin was Suspended in DCM (200 mL) and treated with activated MnO, (62.7g,722 (2S,4S)-1-tert-Butyl 2-(2-(9-((2S,5S)-1-((S)-2- mmol). After stirring for 15 hat RT, the reaction mixture was 50 (methoxycarbonylamino)-3-methylbutanoyl)-5-me filtered over celite and the filter cake was rinsed several times thylpyrrolidine-2-carbonyloxy)-8-oxo-8.9.10.11 with DCM. The combined filtrate (~400 mL) was treated with tetrahydro-5H-dibenzoc.gchromen-3-yl)-2- MeOH (~100 mL) and the mixture was gradually concen oxoethyl) 4-(methoxymethyl)pyrrolidine-1,2- trated under reduced pressure, causing solid material to pre dicarboxylate cipitate from solution. When the liquid volume reached -200 55 mL, the solid was filtered off and rinsed with MeOH. The (2S,4S)-2-(2-(9-Bromo-8-oxo-8,9,10,11-tetrahydro-5H concentration/precipitation/filtration/rinsing sequence was dibenzoc.gchromen-3-yl)-2-oxoethyl) 1-tert-butyl performed 2x more, resulting in the collection of 3 crops of 4-(methoxymethyl)pyrrolidine-1,2-dicarboxylate (8.76 g. powdered 3-(2-bromoacetyl)-10,11-dihydro-5H-dibenzoc, 13.94 mmol) was treated with a solution of (2S,5S)-1-((S)-2- gchromen-8(9H)-one (7.49 g, 56% over 2 steps). 60 (methoxycarbonylamino)-3-methylbutanoyl)-5-methylpyr rolidine-2-carboxylic acid (6.85g, 23.92 mmol) in 2-Me (2S,4S)-1-tert-Butyl 2-(2-oxo-2-(8-oxo-8.9.10.11 THF (70 mL) and CsCO, (3.63 g, 11.15 mmol). The stirred tetrahydro-5H-dibenzoc.gchromen-3-yl)ethyl) reaction mixture was heated to 50° C. for 20h, then cooled to 4-(methoxymethyl)pyrrolidine-1,2-dicarboxylate RT and diluted with EtOAc. The organic phase was washed 65 with HO and brine, then dried over MgSO filtered and 3-(2-Bromoacetyl)-10,11-dihydro-5H-dibenzoc.g concentrated under reduced pressure. The crude residue was chromen-8(9H)-one (7.47 g. 20.1 mmol) and (2S,4S)-1-(tert purified by silica column chromatography (0% to 30%