OA17631A - Polycyclic-carbamoylpyridone compounds and their pharmaceutical use. - Google Patents

Polycyclic-carbamoylpyridone compounds and their pharmaceutical use. Download PDF

Info

Publication number
OA17631A
OA17631A OA1201500240 OA17631A OA 17631 A OA17631 A OA 17631A OA 1201500240 OA1201500240 OA 1201500240 OA 17631 A OA17631 A OA 17631A
Authority
OA
OAPI
Prior art keywords
compound
mmol
hiv
hydrogen
reaction mixture
Prior art date
Application number
OA1201500240
Inventor
Haolun Jin
Teresa Alejandra Trejo Martin
Scott E. Lazerwith
Elizabeth M. Bacon
Jeromy J. Cottell
Zhenhong R. Cai
Hyung-Jung Pyun
Philip Anthony Morganelli
Mingzhe Jl
James G. Taylor
Xiaowu Chen
Michael R. Mish
Manoj C. Desai
Original Assignee
Gilead Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Publication of OA17631A publication Critical patent/OA17631A/en

Links

Abstract

Compounds for use in the treatment of human immunodeficiency virus (HIV) infection are disclosed. The compounds have the following Formula (I) :

Description

Compounds for use in the treatment of human immunodeficiency virus (HIV) infection are disclosed. The compounds hâve the following Formula (I) :
including stereoisomers and pharmaceutically acceptable salts thereof, wherein R1, X, W, Y1, Y2, Z , and Z4 are as defined herein. Methods associated with préparation and use of such compounds, as well as pharmaceutical compositions comprising such compounds, are also disclosed.
O.A.P.I. - B.P. 887, YAOUNDE (Cameroun) - Tel. (237) 222 20 57 00- Fax: (237) 222 20 57 27- Site web: http:/www.oapi.int - Email: oapi@oapi.int
POLYCYCLIC-CARBAMOYLPYRIDONE COMPOUNDS AND THEIR PHARMACEUTICAL USE
CROSS-REFERENCE TO RELATED APPLICATIONS
This patent application claims the benefit under 35 U.S.C. §119(e) ofU.S. Provisional Patent Application No. 61/745,375, filed December 21, 2012, U.S. Provisional Patent Application No. 61/788,397, filed March 15, 2013, and U.S. Provisional Patent Application No. 61/845,803, filed July 12, 2013. The foregoing applications are incorporated herein by reference in their entireties.
BACKGROUND
Field
Compounds, compositions, and methods for the treatment of human immunodeficiency virus (HIV) infection are disclosed. In particular, novel polycyclic carbamoylpyridone compounds and methods for their préparation and use as therapeutic or prophylactic agents are disclosed.
Description of the Related Art
Human immunodeficiency virus infection and related diseases are a major public health problem worldwide. Human immunodeficiency virus type 1 (HIV-1) encodes three enzymes which are required for viral réplication: reverse transcriptase, protease, and integrase. Although drugs targeting reverse transcriptase and protease are in wide use and hâve shown effectiveness, particularly when employed in combination, toxicity and development of résistant strains hâve limited their usefulness (Palella, et al. N. Engl. J Med. (1998) 338:853-860; Richman, D. D. Nature (2001) 410:995-1001).
Pregnane X receptor (PXR) is a nuclear receptor that is one of the key regulators of enzymes involved in metabolism and élimination of small molécules from the body. Activation of PXR is known to up-regulate or induce the production of metabolic enzymes such as cytochrome P450 3A4 (CYP3A4) as well as enzymes involved in transport such as Ο ATP2 in the liver and intestine (Endocrine Reviews (2002) 23(5):687-702). When one drug causes the up-regulation of these and other enzymes by activation of PXR, this can reduce the absorption and/or exposure of a co-administered drug that is susceptible to the up-regulated enzymes. To minimize the risk of this type of drug-drug interaction, it is désirable to minimize PXR activation. Further, it is known that PXR is activated by many different classes of molécules (Endocrine Reviews (2002) 23(5):687-702). Thus for drugs that will be co-administered with other drugs, it is important to test for and minimize PXR activation.
Transporters hâve been identified as playing a rôle in the pharmacokinetic, safety and efficacy profile or drugs, and certain drug-drug interactions are mediated by transporters. See, Giacomini KM, et al. ““Membrane transporters in drug development,” Nat.Rev Drug Discov. 9: 215-236, 2010; Zhang L, et al. “Transporter-Mediated Drug-Drug Interactions,” Clin. Pharm. Ther. 89(4):481-484 (2011). -One tranporter, the organic cation transporter 2 (OCT2; SLC22A2), is a member of the soluté carrier (SLC) super-family of transporters and is primarily localized on the basolateral membrane of the rénal proximal tubule. OCT2, in concert with apical expressed multidrug and toxin extrusion (MATE) transporters 1 and 2-K, is believed to form the major cationic sécrétion pathway in the kidney and has been shown to transport endogenous compounds including créatinine and xenobiotics including metformin. Inhibition of OCT2 can thus lead to increased levels of sérum créatinine and the potential for increased levels of other OCT2 substrates. It is important as well to test and reduce OCT2 inhibition of drugs.
A goal of antirétroviral therapy is to achieve viral suppression in the HIV infected patient. Treatment guidelines published by the United States Department of Health and Human Services provide that achievement of viral suppression requires the use of combination thérapies, i.e., several drugs from at least two or more drug classes. (Panel on Antirétroviral Guidelines for Adults and Adolescents. Guidelines for the use of antirétroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services. Available at http://aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf Section accessed March 14, 2013.) In addition, decisions regarding the treatment of HIV infected patients are complicated when the patient requires treatment for other medical conditions (Id. at E-12). Because the standard of care requires the use of multiple different drugs to suppress HIV, as well as to treat other conditions the patient may be experiencing, the potential for drug interaction is a criterion for sélection of a drug regimen. As such, there is a need for antirétroviral thérapies having a decreased potential for drug interactions.
Accordingly, there is a need for new agents that inhibit the réplication of HIV and that minimize PXR activation when co-administered with other drugs.
BRIEF SUMMARY
The présent invention is directed to novel polycyclic carbamoylpyridone compounds, having antiviral activity, including stereoisomers and pharmaceutically acceptable salts thereof, and the use of such compounds in the treatment of HEV infections. The compounds of the invention may be used to inhibit the activity of HIV integrase and may be used to reduce HIV réplication.
In one embodiment of the présent invention, compounds having the following Formula (I) are provided:
or a stereoisomer or pharmaceutically acceptable sait thereof, wherein:
X is -O- or -NZ3- or -CHZ3-;
W is -CHZ2-;
Z1, Z2 and Z3 are each, independently, hydrogen or Ci-3alkyl, or wherein Z1 and Z2 or Z1 and Z3, taken together, form -L- wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, or -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, wherein at least one of Z1 and Z2 or Z1 and Z3, taken together, form -L-;
Z4 is a bond, -CH2-, or -CH2CH2-;
Y1 and Y2 are each, independently, hydrogen, Ci-3alkyl or Ci-3haloalkyl;
R1 is phenyl substituted with one to three halogens; and each Ra is, independently, hydrogen, halo, hydroxyl or Ci-4alkyl.
In another embodiment of the présent invention, compounds having the following Formula (I) are provided:
or a stereoisomer or pharmaceutically acceptable sait thereof, wherein:
X is -O- or -NZ3- or -CHZ3-;
W is -O- or -NZ2- or -CHZ2-;
Z1, Z2 and Z3 are each, independently, hydrogen or Ci-3alkyl, or wherein Z1 and Z2 or Z1 and Z3, taken together, form -L- wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2 OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2, -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C( Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2C(R a)2-, -C(Ra)2C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SO2C(Ra)2-, -C (Ra)2SO2NRaC(Ra)2- or -C(Ra)2NRaSO2C(Ra)2-;
Z4 is a bond or -CH2-, -CH2CH2-, -CH2CH2CH2-, -CH2OCH2-, -CH2NRaCH2-, -CH2SCH2-,-CH2S(O )CH2- or -CH2SO2CH2-;
Y1 and Y2 are each, independently, hydrogen, Ci-3alkyl or Ci-3haloalkyl, or Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms, wherein the carbocyclic or heterocyclic ring is optionally substituted with one or more Ra;
R1 is optionally substituted aiyl or optionally substituted heteroaryl; and each Ra is, independently, hydrogen, halo, hydroxyl or Cwalkyl, or wherein two Ra groups, together with the carbon atom to which they are attached, form =0, and wherein at least one of: (i) Z1 and Z2 or Z1 and Z3, taken together, form -L-; or (ii) Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
In another embodiment, a pharmaceutical composition is provided comprising a compound having Formula (I), or a stereoisomer or pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
The invention also provides the use of a pharmaceutical composition as described hereinabove for the treatment of an HIV infection in a human being having or at risk of having the infection.
In another embodiment, a method of using a compound having Formula (I) in therapy is provided. In particular, a method of treating the prolifération of the HIV virus, treating AIDS, or delaying the onset of AIDS or ARC symptoms in a mammal (e.g., a human) is provided, comprising administering to the mammal a compound having Formula (I), or a stereoisomer or pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
In another embodiment, use of a compound of Formula (I) as described herein, or a pharmaceutically acceptable sait thereof, for the treatment of an HIV infection in a human being having or at risk of having the infection is disclosed.
In another embodiment, the use of a compound of Formula (I) as described herein, or a pharmaceutically acceptable sait thereof, for the manufacture of a médicament for the treatment of an HIV infection in a human being having or at risk of having the infection is disclosed.
In another embodiment, an article of manufacture comprising a composition effective to treat an HIV infection; and packaging material comprising a label which indicates that the composition can be used to treat infection by HIV is disclosed. Exemplary compositions comprise a compound of Formula (I) according to this invention or a pharmaceutically acceptable sait thereof.
In still another embodiment, a method of inhibiting the réplication ofHIV is disclosed. The method comprises exposing the virus to an effective amount of the compound of Formula (I), or a sait thereof, under conditions where réplication ofHIV is inhibited.
In another embodiment, the use of a compound of Formula (I) to inhibit the activity of the HIV integrase enzyme is disclosed.
In another embodiment, the use of a compound of Formula (I), or a sait thereof, to inhibit the réplication ofHIV is disclosed.
Other embodiments, objects, features and advantages will be set forth in the detailed description of the embodiments that follows, and in part will be apparent ffom the description, or may be learned by practice, of the claimed invention. These objects and advantages will be realized and attained by the processes and compositions particularly pointed out in the written description and claims hereof. The foregoing Summary has been made with the understanding that it is to be considered as a brief and general synopsis of some of the embodiments disclosed herein, is provided solely for the benefit and convenience of the reader, and is not intended to limit in any manner the scope, or range of équivalents, to which the appended claims are lawfully entitled.
DETAILED DESCRIPTION
In the following description, certain spécifie details are set forth in order to provide a thorough understanding of various embodiments of the invention. However, one skilled in the art will understand that the invention may be practiced without these details. The description below of several embodiments is made with the understanding that the présent disclosure is to be considered as an exemplification of the claimed subject matter, and is not intended to limit the appended claims to the spécifie embodiments illustrated. The headings used throughout this disclosure are provided for convenience only and are not to be construed to limit the claims in any way. Embodiments illustrated under any heading may be combined with embodiments illustrated under any other heading.
Définitions
Unless the context requires otherwise, throughout the présent spécification and claims, the word “comprise” and variations thereof, such as, “comprises” and “comprising” are to be construed in an open, inclusive sense, that is as “including, but not limited to”.
Reference throughout this spécification to “one embodiment” or “an embodiment” means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the présent invention. Thus, the appearances of the phrases “in one embodiment” or “in an embodiment” in various places throughout this spécification are not necessarily ail referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.
Unless the context requires otherwise, reference throughout this spécification to “a compound of Formula (1)” or “compounds of Formula (1)” refers to ail embodiments of Formula (I), including, for example, compounds of Formulas (II-A), (II-B), (Π-C), (ΙΠ-Α), (ΙΠ-Β), (ΠΙ-C), (ΠΙ-D), (ΠΙ-Ε), (ΠΙ-F), (ΠΙ-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as well as the spécifie compounds disclosed herein.
“Amino” refers to the -NFfo radical.
“Cyano” refers to the -CN radical.
“Hydroxy” or “hydroxyl” refers to the -OH radical.
“Imino” refers to the =NH substituent.
“Nitro” refers to the -NO2 radical.
“Oxo” refers to the =0 substituent.
“Thioxo” refers to the =S substituent.
“Alkyl” refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), having from one to twelve carbon atoms (C1-C12 alkyl), preferably one to eight carbon atoms (Ci-Cg alkyl) or one to six carbon atoms (Ci-Ce alkyl), and which is attached to the rest of the molécule by a single bond, e.g., methyl, ethyl, w-propyl, 1-methylethyl (Ao-propyl), n-butyl, w-pentyl,
1,1-dimethylethyl (Abutyl), 3-methylhexyl, 2-methylhexyl, ethenyl, prop-1-enyl, but-1-enyl, pent-1-enyl, penta-l,4-dienyl, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the spécification, an alkyl group may be optionally substituted.
“Alkylene” or “alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molécule to a radical group, consisting solely of carbon and hydrogen, which is saturated or unsaturated (i.e., contains one or more double and/or triple bonds), and having from one to twelve carbon atoms, e.g., methylene, ethylene, propylene, n-butylene, ethenylene, propenylene, n-butenylene, propynylene, H-butynylene, and the like. The alkylene chain is attached to the rest of the molécule through a single or double bond and to the radical group through a single or double bond. The points of attachment of the alkylene chain to the rest of the molécule and to the radical group can be through one carbon or any two carbons within the chain. Unless stated otherwise specifically in the spécification, an alkylene chain may be optionally substituted.
“Alkoxy” refers to a radical of the formula -ORa where Ra is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the spécification, an alkoxy group may be optionally substituted.
“Alkylamino” refers to a radical of the formula -NHRa or -NRaRa where each Ra is, independently, an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the spécification, an alkylamino group may be optionally substituted.
Thioalkyl” refers to a radical of the formula -SRa where Ra is an alkyl radical as defined above containing one to twelve carbon atoms. Unless stated otherwise specifically in the spécification, a thioalkyl group may be optionally substituted.
“Aryl” refers to a monocylic hydrocarbon ring System radical comprising hydrogen and 6 to 18 carbon atoms. Aryl radicals include, but are not limited to, aryl radicals derived from benzene. Unless stated otherwise specifically in the spécification, the term “aryl” or the prefix “ar-“ (such as in “aralkyl”) is meant to include aryl radicals that are optionally substituted.
“Aralkyl” refers to a radical of the formula -Rb-Rc where Rb is an alkylene chain as defined above and Rc is one or more aryl radicals as defined above, for example, benzyl. Unless stated otherwise specifically in the spécification, an aralkyl group may be optionally substituted.
“Cycloalkyl” or “carbocyclic ring” refers to a stable non-aromatic monocyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, having from three to fifteen carbon atoms, preferably having from three to ten carbon atoms, and which is saturated or unsaturated and attached to the rest of the molécule by a single bond. Monocyclic radicals include, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Unless otherwise stated specifically in the spécification, a cycloalkyl group may be optionally substituted.
“Cycloalkylalkyl” refers to a radical of the formula -RbRd where Rb is an alkylene chain as defined above and Rd is a cycloalkyl radical as defined above. Unless stated otherwise specifically in the spécification, a cycloalkylalkyl group may be optionally substituted.
“Halo” or “halogen” refers to bromo, chloro, fluoro or iodo.
“Haloalkyl” refers to an alkyl radical, as defîned above, that is substituted by one or more halo radicals, as defined above, e.g., trifluoromethyl, difluoromethyl, trichloromethyl, 2,2,2-trifluoroethyl, 1,2-difluoroethyl, 3-bromo-2-fluoropropyl, 1,2-dibromoethyl, and the like. Unless stated otherwise specifically in the spécification, a haloalkyl group may be optionally substituted.
“Heterocyclyl” or “heterocyclic ring” refers to a stable 3- to 18-membered non-aromatic ring radical which consists of two to twelve carbon atoms and from one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. In the embodiments disclosed herein, the heterocyclyl radical is a monocyclic ring system; and the heterocyclyl radical may be partially or fully saturated. Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl, [l,3]dithianyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the spécification, a heterocyclyl group may be optionally substituted.
“/V-heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molécule is through a nitrogen atom in the heterocyclyl radical. Unless stated otherwise specifically in the spécification, an TV-heterocyclyl group may be optionally substituted.
“Heterocyclylalkyl” refers to a radical of the formula -RbRe where Rb is an alkylene chain as defined above and Re is a heterocyclyl radical as defined above, and if the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl may be attached to the alkyl radical at the nitrogen atom. Unless stated otherwise specifically in the spécification, a heterocyclylalkyl group may be optionally substituted.
“Heteroaryl” refers to a 5- to 14-membered monocyclic ring system radical comprising hydrogen atoms, one to thirteen carbon atoms, one to six heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. Examples include, but are not limited to, azepinyl, furanyl, furanonyl, isothiazolyl, imidazolyl, isoxazolyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 1-oxidopyridinyl, 1-oxidopyrimidinyl, 1oxidopyrazinyl, 1-oxidopyridazinyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, thiophenyl, and thienyl. Unless stated otherwise specifically in the spécification, a heteroaryl group may be optionally substituted.
“Λ-heteroaryl” refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molécule is through a nitrogen atom in the heteroaryl radical. Unless stated otherwise specifically in the spécification, an /V-heteroaryl group may be optionally substituted.
“Heteroarylalkyl” refers to a radical of the formula -RbRf where Rb is an alkylene chain as defined above and Rf is a heteroaryl radical as defined above. Unless stated otherwise specifically in the spécification, a heteroarylalkyl group may be optionally substituted.The term “substituted” used herein means any of the above groups (i.e., alkyl, alkylene, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, TV-heterocyclyl, heterocyclylalkyl, heteroaryl, Λ-heteroaryl and/or heteroarylalkyl) wherein at least one hydrogen atom is replaced by a bond to a non-hydrogen atoms such as, but not limited to: a halogen atom such as F, Cl,
Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, and ester groups; a sulfur atom in groups such as thiol groups, thioalkyl groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as trialkylsilyl groups, dialkylarylsilyl groups, alkyldiarylsilyl groups, and triarylsilyl groups; and other heteroatoms in various other groups. “Substituted” also means any of the above groups in which one or more hydrogen atoms are replaced by a higher-order bond (e.g., a doubleor triple-bond) to a heteroatom such as oxygen in oxo, carbonyl, carboxyl, and ester groups; and nitrogen in groups such as imines, oximes, hydrazones, and nitriles. For example, “substituted” includes any of the above groups in which one or more hydrogen atoms are replaced with NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, -NRgC^NRQNRgRh, NRgSO2Rh, -OC(=O)NRgRh, -ORg, -SRg, -SORg, -SO2Rg, -OSO2Rg, -SO2ORg, =NSO2Rg, and -SO2NRgRh. “Substituted also means any of the above groups in which one or more hydrogen atoms are replaced with -C(=O)Rg, -C(=O)ORg, -C(=O)NRgRh, -CH2SO2Rg, -CH2SO2NRgRh. In the foregoing, Rg and Rh are the same or different and independently hydrogen, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, JV-heterocyclyl, heterocyclylalkyl, heteroaryl, TV-heteroaryl and/or heteroarylalkyl. “Substituted” further means any of the above groups in which one or more hydrogen atoms are replaced by a bond to an amino, cyano, hydroxyl, imino, nitro, oxo, thioxo, halo, alkyl, alkoxy, alkylamino, thioalkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl, haloalkyl, heterocyclyl, JV-heterocyclyl, heterocyclylalkyl, heteroaryl, M-heteroaryl and/or heteroarylalkyl group. In addition, each of the foregoing substituents may also be optionally substituted with one or more of the above substituents.
The term “protecting group,” as used herein, refers to a labile chemical moiety which is known in the art to protect reactive groups including without limitation, hydroxyl and amino groups, against undesired reactions during synthetic procedures. Hydroxyl and amino groups protected with a protecting group are referred to herein as “protected hydroxyl groups” and “protected amino groups”, respectively. Protecting groups are typically used selectively and/or orthogonally to protect sites during reactions at other reactive sites and can then be removed to leave the unprotected group as is or available for further reactions. Protecting groups as known in the art are described generally in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd édition, John Wiley & Sons, New York (1999). Generally, groups are protected or présent as a precursor that will be inert to reactions that modify other areas of the parent molécule for conversion into their final groups at an appropriate time. Further représentative protecting or precursor groups are discussed in Agrawal, et al., Protocols for Oligonucleotide Conjugates, Eds, Humana Press; New Jersey, 1994; Vol. 26 pp. 1-72. Examples of “hydroxyl protecting groups” include, but are not limited to, t-butyl, t-butoxymethyl, methoxy methyl, tetrahydropyranyl, 1-ethoxyethyl, l-(2-chloroethoxy)ethyl, 2trimethylsilylethyl, p-chlorophenyl, 2,4-dinitrophenyl, benzyl, 2,6-dichlorobenzyl, diphenylmethyl, p-nitrobenzyl, triphenylmethyl, trimethylsilyl, triethylsilyl, tbutyldimethylsilyl, t-butyldiphenylsilyl (TBDPS), triphenylsilyl, benzoylformate, acetate, chloroacetate, trichloroacetate, trifluoroacetate, pivaloate, benzoate, pphenylbenzoate, 9-fluorenylmethyl carbonate, mesylate and tosylate. Examples of “amino protecting groups” include, but are not limited to, carbamate-protecting groups, such as 2-trimethylsilylethoxy carbonyl (Teoc), 1 -methyl- l-(4-biphenylyl)ethoxycarbonyl (Bpoc), t-butoxycarbonyl (BOC), allyloxycarbonyl (Alloc), 9fluorenylmethyloxycarbonyl (Fmoc), and benzyloxycarbonyl (Cbz); amide protecting groups, such as formyl, acetyl, trihaloacetyl, benzoyl, and nitrophenylacetyl; sulfonamide-protecting groups, such as 2-nitrobenzenesulfonyl; and imine and cyclic imide protecting groups, such as phthalimido and dithiasuccinoyl.
The invention disclosed herein is also meant to encompass ail pharmaceutically acceptable compounds of Formula (I) being isotopically-labeled by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H, 3H, nC, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 31P, 32P, 35S, 18F, 36C1, 123I, and 125I, respectively. These radiolabeled compounds could be useful to help détermine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action. Certain isotopically-labeled compounds of Formula (I), for example, those incorporating a radioactive isotope, are usefùl in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of détection.
Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability. For example, in vivo half-life may increase or dosage requirements may be reduced. Thus, heavier isotopes may be preferred in some circumstances.
Substitution with positron emitting isotopes, such as nC, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of Formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
The invention disclosed herein is also meant to encompass the in vivo metabolic products of the disclosed compounds. Such products may resuit from, for example, the oxidation, réduction, hydrolysis, amidation, estérification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, the invention includes compounds produced by a process comprising administering a compound of this invention to a mammal for a period of time sufficient to yield a metabolic product thereof. Such products are typically identified by administering a radiolabeled compound of the invention in a détectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
“Stable compound” and “stable structure” are meant to indicate a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
“Mammal” includes humans and both domestic animais such as laboratory animais and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non-domestic animais such as wildlife and the like.
“Optional” or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, “optionally substituted aryl” means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
“Pharmaceutically acceptable carrier, diluent or excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonie agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animais.
“Pharmaceutically acceptable sait” refers to a sait of a compound that is pharmaceutically acceptable and that possesses (or can be converted to a form that possesses) the desired pharmacological activity of the parent compound. Examples of “pharmaceutically acceptable salts” of the compounds disclosed herein include salts derived from an appropriate base, such as an alkali métal (for example, sodium), an alkaline earth métal (for example, magnésium), ammonium and NXA (wherein X is C1-C4 alkyl). Pharmaceutically acceptable salts of a nitrogen atom or an amino group include for example salts of organic carboxylic acids such as acetic, benzoic, camphorsulfonic, citric, glucoheptonic, gluconic, lactic, fumaric, tartaric, maleic, malonic, malic, mandelic, isethionic, lactobionic, succinic, 2-napththalenesulfonic, oleic, palmitic, propionic, stearic, and trimethylacetic acids; organic sulfonic acids, such as methanesulfonic, ethanesulfonic, benzenesulfonic and p-toluenesulfonic acids; and inorganic acids, such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric and sulfamic acids. Pharmaceutically acceptable salts of a compound of a hydroxy group include the anion of said compound in combination with a suitable cation such as Na+ and NX4+ (wherein X is independently selected from H or a C1-C4 alkyl group). Pharmaceutically acceptable salts also include salts formed when an acidic proton présent in the parent compound is replaced by either a métal ion, e.g., an alkali métal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as diethanolamine, triethanolamine, N-methylglucamine and the like. Also included in this définition are ammonium and substituted or quatemized ammonium salts. Représentative non-limiting lists of pharmaceutically acceptable salts can be found in
S.M. Berge et al., J. Pharma Sci., 66(1), 1-19 (1977), and Remington: The Science and Practice of Pharmacy, R. Hendrickson, ed., 21st édition, Lippincott, Williams & Wilkins, Philadelphia, PA, (2005), at p. 732, Table 38-5, both of which are hereby incorporated by reference herein.
For therapeutic use, salts of active ingrédients of the compounds disclosed herein will typically be pharmaceutically acceptable, i.e. they will be salts derived from a physiologically acceptable acid or base. However, salts of acids or bases which are not pharmaceutically acceptable may also find use, for example, in the préparation or purification of a compound of Formula (I) or another compound of the invention. Ail salts, whether or not derived from a physiologically acceptable acid or base, are within the scope of the présent invention.
Métal salts typically are prepared by reacting the métal hydroxide with a compound of this invention. Examples of métal salts which are prepared in this way are salts containing Li+, Na+, and K+. A less soluble métal sait can be precipitated from the solution of a more soluble sait by addition of the suitable métal compound.
In addition, salts may be formed from acid addition of certain organic and inorganic acids, e.g., HCl, HBr, H2SO4, H3PO4 or organic sulfonic acids, to basic centers, typically amines. Finally, it is to be understood that the compositions herein comprise compounds disclosed herein in their un-ionized, as well as zwitterionic form, and combinations with stoichiometric amounts of water as in hydrates.
Often crystallizations produce a solvaté of the compound of the invention. As used herein, the terni “solvaté” refers to an aggregate that comprises one or more molécules of a compound of the invention with one or more molécules of solvent. The solvent may be water, in which case the solvaté may be a hydrate. Altematively, the solvent may be an organic solvent. Thus, the compounds of the présent invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. The compound of the invention may be true solvatés, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent.
A “pharmaceutical composition” refers to a formulation of a compound of the invention and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans. Such a medium includes ail pharmaceutically acceptable carriers, diluents or excipients therefor.
“Effective amount” or “therapeutically effective amount” refers to an amount of a compound according to the invention, which when administered to a patient in need thereof, is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds hâve utility. Such an amount would be sufficient to elicit the biological or medical response of a tissue system, or patient that is sought by a researcher or clinician. The amount of a compound according to the invention which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological activity, the composition used for administration, the time of administration, the route of administration, the rate of excrétion of the compound, the duration of the treatment, the type of disease-state or disorder being treated and its severity, drugs used in combination with or coincidentally with the compounds of the invention, and the âge, body weight, general health, sex and diet of the patient. Such a therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the state of the art, and this disclosure.
The term treatment as used herein is intended to mean the administration of a compound or composition according to the présent invention to alleviate or eliminate symptoms of HIV infection and/or to reduce viral load in a patient. The term treatment also encompasses the administration of a compound or composition according to the présent invention post-exposure of the individual to the virus but before the appearance of symptoms of the disease, and/or prior to the détection of the virus in the blood, to prevent the appearance of symptoms of the disease and/or to prevent the virus from reaching detectible levels in the blood, and the administration of a compound or composition according to the présent invention to prevent périnatal transmission of HIV from mother to baby, by administration to the mother before giving birth and to the child within the first days of life.
The term antiviral agent as used herein is intended to mean an agent (compound or biological) that is effective to inhibit the formation and/or réplication of a virus in a human being, including but not limited to agents that interfère with either host or viral mechanisms necessary for the formation and/or réplication of a virus in a human being.
The term inhibitor of HIV réplication as used herein is intended to mean an agent capable of reducing or eliminating the ability of HIV to replicate in a host cell, whether in vitro, ex vivo or in vivo.
The compounds of the invention, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (5)- or, as (D)- or (L)- for amino acids. The présent invention is meant to include ail such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a sait or dérivative) using, for example, chiral high pressure liquid chromatography (HPLC). When the compounds described herein contain olefmic double bonds or other centres of géométrie asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z géométrie isomers. Likewise, ail tautomeric forms are also intended to be included.
A “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The présent invention contemplâtes various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molécules are nonsuperimposeable mirror images of one another.
A “tautomer” refers to a proton shift from one atom of a molécule to another atom of the same molécule. The présent invention includes tautomers of any said compounds.
A “prodrug” refers to a compound that is chemically designed to efficiently liberate the parent drug after overcoming biological barriers to oral delivery. In certain embodiments, the présent invention includes prodrugs of the compounds of Formula (I).
Compounds
As noted above, in one embodiment of the présent invention, compounds having antiviral activity are provided, the compounds having the following Formula (I):
or a stereoisomer or pharmaceutically acceptable sait thereof, wherein:
X is -O- or -NZ3- or -CHZ3-;
W is -CHZ2-;
Z1, Z2 and Z3 are each, independently, hydrogen or Ci-3alkyl, or wherein
Z1 and Z2 or Z1 and Z3, taken together, form -L- wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, or -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, wherein at least one of Z1 and Z2 or Z1 and Z3, taken together, form -L-;
Z4 is a bond, -CH2-, or -CH2CH2-;
Y1 and Y2 are each, independently, hydrogen, Ci_3alkyl or Crohaloalkyl;
R1 is phenyl substituted with one to three halogens; and each Ra is, independently, hydrogen, halo, hydroxyl or Ci-4alkyl.
In another embodiment, compounds are provided having the following
Formula (II-A):
R1
In another embodiment, compounds are provided having the following
Formula (II-B):
In another embodiment, compounds are provided having the following
Formula (II-C):
(Π-C)
In another embodiment, L is -C(Ra)2-. In a further embodiment, L is -C(Ra)2C(Ra)2-. In still a fùrther embodiment, L is -C(Ra)2C(Ra)2C(Ra)2-. In still a further embodiment, each Ra is hydrogen. In still a further embodiment, one Ra is methyl and each remaining Ra is hydrogen. In still a fùrther embodiment, one Ra is halogen and each remaining Ra is hydrogen. In still a further embodiment, two Ra are halogen and each remaining Ra is hydrogen. In still a further embodiment, one Ra is halogen and each remaining Ra is hydrogen.
In another embodiment, X is -O-. In another embodiment, X is -NZ3-. In another embodiment, X is -NH-. 16. In another embodiment, X is -CHZ3- and Z1 and Z3, taken together, form -L-. In a fùrther embodiment, Z2 is hydrogen. In another embodiment, X is -CHz-.
In another embodiment, Z4 is a bond or -CH2-. In another embodiment, Z4 is -CH2-. In another embodiment, Z4 is a bond.
In another embodiment, Y1 and Y2 are each independently hydrogen, methyl or trifluoromethyl.
In another embodiment, R1 is substituted with one halogen. In a further embodiment, R1 is 4-fluorophenyl or 2-fluorophenyl.
In another embodiment, R1 is substituted with two halogens. In a further embodiment, R1 is 2,4-difluorophenyl, 2,3-difluorophenyl, 2,6-difluorophenyl, 3-fluoro4-chlorophenyl, 3,4-difluorophenyl, 2-fluoro-4-chlorophenyl, or 3,5-difluorophenyl. In still a further embodiment, R1 is 2,4-difluorophenyl.
In another embodiment, R1 is substituted with three halogens. In a fùrther embodiment, R1 is 2,4,6-trifluorophenyI or 2,3,4-trifluorophenyl. In still a fùrther embodiment, R1 is 2,4,6-trifluorophenyl.
In one embodiment, a pharmaceutical composition is provided comprising a compound of any one of the Formulas (I), (II-A), (Π-B), or (Π-C), as noted above, or a stereoisomer or pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
Another embodiment is provided comprising a method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of the Formulas (I), (II-A), (Π-B), or (II-C), as noted above, or a pharmaceutical composition thereof. Another embodiment is provided comprising a method of treating or preventing an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of the Formulas (I), (II-A), (Π-B), or (II-C), as noted above, or a pharmaceutical composition thereof.
In another embodiment, the use of a compound of any one of the Formulas (I), (II-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for the treatment of an HIV infection in a human having or at risk of having the infection is provided. In another embodiment, the use of a compound of any one of the Formulas (I), (II-A), (Π-B), or (Π-C), as noted above, or a pharmaceutical composition thereof, for the treatment or prévention of an HIV infection in a human having or at risk of having the infection is provided.
In another embodiment, the use in medical therapy of a compound of any one of the Formulas (I), (Π-A), (Π-B), or (Π-C), as noted above, or a pharmaceutical composition thereof, is provided.
In another embodiment, the use of a compound of any one of the Formulas (I), (Π-A), (II-B), or (II-C), as noted above, or a pharmaceutical composition thereof, for use in the therapeutic treatment of an HIV infection is provided. In another embodiment, the use of a compound of any one of the Formulas (I), (II-A), (Ïï-B), or (Π-C), as noted above, or a pharmaceutical composition thereof, for use in the prophylactic or therapeutic treatment of an HIV infection is provided.
As further noted above, in another embodiment of the présent invention, compounds having antiviral activity are provided, the compounds having the following Formula (I):
or a stereoisomer or pharmaceutically acceptable sait thereof, wherein:
X is -O- or -NZ3- or -CHZ3-;
W is -O- or -NZ2- or -CHZ2-;
Z1, Z2 and Z3 are each, independently, hydrogen, Ci-3alkyl or Ci-3haloalkyl, or wherein Z1 and Z2 or Z1 and Z3, taken together, form -L- wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2 OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2, -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C( Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2C(R a)2-, -C(Ra)2C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SO2C(Ra)2-, -C (Ra)2SO2NRaC(Ra)2- or -C(Ra)2NRaSO2C(Ra)2-;
Z4 is a bond or -CH2-, -CH2CH2-, -CH2CH2CH2-, -CH2OCH2-, -CH2NRaCH2-, -CH2SCH2-,-CH2S(O )CH2- or-CH2SO2CH2-;
Y1 and Y2 are each, independently, hydrogen or Ci-3alkyl, or Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms, wherein the carbocyclic or heterocyclic ring is optionally substituted with one or more Ra;
R1 is optionally substituted aryl or optionally substituted heteroaryl; and each Ra is, independently, hydrogen, halo, hydroxyl or Ci^alkyl, or wherein two Ra groups, together with the carbon atom to which they are attached, form =0, and wherein at least one of: (i) Z1 and Z2 or Z1 and Z3, taken together, form -L-; or (ii) Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
In another embodiment, W is -CHZ2-.
In another embodiment, Z1 and Z2 or Z1 and Z3, taken together, form -L-.
In another embodiment, compounds are provided having one of the following Formulas (II-A), (Π-B), or (Π-C):
R1 ; or
(II-C) wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2 OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)210 , -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C(
Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2C(R a)2-, -C(Ra)2C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SO2C(Ra)2-, -C (Ra)2SO2NRaC(Ra)2- or -C(Ra)2NRaSO2C(Ra)2-.
In another embodiment, Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
In another embodiment, compounds are provided having one of the following Formulas (ΙΠ-Α), (ΠΙ-Β), (ΠΙ-C) or (ΠΙ-D):
Z1 O OH (III-A)
(III-B)
(ΙΠ-C)
(III-D) wherein Z1 and Z3 are each, independently, hydrogen or Ci-3alkyl.
In another embodiment, compounds are provided having one of the following Formulas (III-E), (ΠΙ-F), (ΙΠ-G) or (III-H):
Z1 O OH (III-E)
(III-F)
(ΙΠ-Η) wherein Z1 and Z3 are each, independently, hydrogen or Ci-3alkyl.
In another embodiment, both (i) Z1 and Z2 or Z1 and Z3, taken together, form -L-, and (ii) Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
In another embodiment, compounds are provided having one of the following Formulas (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG) or (IV-AH):
(IV-AA)
O OH (IV-AB)
O OH (IV-AC)
O OH (IV-AD)
O OH (IV-AE)
; or
(IV-AH) wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2- -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2
OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)210 , -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C(
Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2C(R a)2-, -C(Ra)2C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SO2C(Ra)2-, -C (Ra)2SO2NRaC(Ra)2- or -C(Ra)2NRaSO2C(Ra)2-,
In another embodiment, compounds are provided having one of the following Formulas (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG) or (IV-BH):
O OH (IV-BA)
O OH (IV-BB)
O OH (IV-BC)
O OH (IV-BD)
O OH (IV-BE)
O OH (IV-BF)
; or
wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2 OC(Ra)2-, -C(Ra)2NRaC(Ra)2- -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2, -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C( Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2C(R a)2-, -C(Ra)2C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SO2C(Ra)2-, -C (Ra)2SO2NRaC(Ra)2- or -C(Ra)2NRaSO2C(Ra)2-,
In another embodiment, L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, or -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, In a further embodiment, L is -C(Ra)2-, In still a further embodiment, L is -C(Ra)2C(Ra)2-, In still a further embodiment, L is -C(Ra)2C(Ra)2C(Ra)2-, In still a further embodiment, each Ra is hydrogen. In still a further embodiment, one Ra is methyl and each remaining Ra is hydrogen. In still a further embodiment, one Ra is halogen and each remaining Ra is hydrogen. In still a further embodiment, two Ra are halogen and each remaining Ra is hydrogen. In still a further embodiment, one Ra is halogen and each remaining Ra is hydrogen.
In another embodiment, L is -C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, or -C(Ra)2SO2C(Ra)2-. In a fùrther embodiment, L is -C(Ra)2OC(Ra)2-. In still a further embodiment, each Ra is hydrogen. In still a fùrther embodiment, one Ra is methyl and each remaining Ra is hydrogen. In still a further embodiment, one Ra is halogen and each remaining Ra is hydrogen. In still a further embodiment, two Ra are halogen and each remaining Ra is hydrogen. In still a fùrther embodiment, one Ra is halogen and each remaining Ra is hydrogen.
In another embodiment, X is -O-. In a further embodiment, Z2 is hydrogen. In another embodiment, X is -NZ3-. In another embodiment, X is -NH-. In another embodiment, X is -CHZ3-. In another embodiment, X is -CH2-.
In another embodiment, Z4 is a bond or -CH2-. In another embodiment, Z4 is -CH2-. In another embodiment, Z4 is a bond.
In another embodiment, Y1 and Y2 are each independently hydrogen, methyl or trifluoromethyl.
In another embodiment, R1 is substituted with one halogen. In a further embodiment, R1 is 4-fluorophenyl or 2-fluorophenyl.
In another embodiment, R1 is phenyl. In another embodiment, R1 is pyridinyl.
In another embodiment, R1 is substituted with at least one halogen.
In another embodiment, R1 is substituted with one halogen. In a further embodiment, R1 is 4-fluorophenyl or 2-fluorophenyl.
In another embodiment, R1 is substituted with two halogens. In a fùrther embodiment, R1 is 2,4-difluorophenyl, 2,3-difluorophenyl, 2,6-difluorophenyl, 3-fluoro4-chlorophenyl, 3,4-difluorophenyl, 2-fluoro-4-chlorophenyl, or 3,5-difluorophenyl. In still a further embodiment, R1 is 2,4-difluorophenyl.
In another embodiment, R1 is substituted with three halogens. In a fùrther embodiment, R1 is 2,4,6-trifluorophenyl or 2,3,4-trifluorophenyl. In still a fùrther embodiment, R1 is 2,4,6-trifluorophenyl.
In another embodiment, R1 is 3-trifluoromethyl-4-fluorophenyl or 2cyclopropoxy-4-fluorophenyl.
In one embodiment, a pharmaceutical composition is provided comprising a compound of any one of Formulas (I), (Π-A), (II-B), (II-C), (III-A), (ΙΠ-Β), (III-C), (III-D), (III-E), (III-F), (ΙΠ-G), (ΙΠ-Η), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a stereoisomer or pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
Another embodiment is provided comprising a method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of Formulas (I), (II-A), (II-B), (II-C), (ΠΙ-Α), (ΙΠ-Β), (ΙΠ-C), (ΠΙ-D), (ΠΙ-E), (III-F), (ΠΙ-G), (III-H), (IVAA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IVBB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof. Another embodiment is provided comprising a method of treating or preventing an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of Formulas (I), (Π-A), (II-B), (II-C), (III-A), (III-B), (III-C), (III-D), (III-E), (III-F), (ΙΠ-G), (ΙΠ-Η), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof.
In another embodiment, the use of a compound of any one of Formulas (I), (II-A), (II-B), (II-C), (ΠΙ-Α), (III-B), (ΠΙ-C), (III-D), (ΠΙ-E), (III-F), (III-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof for the treatment of an HIV infection in a human having or at risk of having the infection. In another embodiment, the use of a compound of any one of Formulas (I), (II-A), (Π-B), (II-C), (ΠΙ-Α), (III-B), (III-C), (III-D), (ΠΙ-Ε), (III-F), (ΠΙ-G), (ΙΠ-Η), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof for the treatment or prévention of an HIV infection in a human having or at risk of having the infection.
In another embodiment, the use in medical therapy of a compound of any one of the Formulas (I), (Π-A), (H-B), (II-C), (ΙΠ-Α), (ΙΠ-Β), (ΙΠ-C), (ΙΠ-D), (ΠΙ-Ε), (III-F), (ΠΙ-G), (ΙΠ-Η), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof, is provided.
In another embodiment, the use of a compound of any one ofthe Formulas (I), (II-A), (II-B), (II-C), (ΠΙ-Α), (III-B), (III-C), (ΠΙ-D), (ΠΙ-Ε), (ΠΙ-F), (ΠΙ-G), (ΙΠ-Η), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof, for use in the therapeutic treatment of an HIV infection is provided. In another embodiment, the use of a compound of any one of the Formulas (I), (Π-A), (II-B), (Π-C), (III-A), (III-B), (III-C), (ΙΠ-D), (ΙΠ-Ε), (III-F), (ΙΠ-G), (III-H), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (TV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as noted above, or a pharmaceutical composition thereof, for use in the prophylactic or therapeutic treatment of an HIV infection is provided.
It is understood that any embodiment of the compounds of Formulas (I), (II-A), (II-B), (II-C), (ΠΙ-Α), (III-B), (ΙΠ-C), (ΠΙ-D), (ΠΙ-E), (III-F), (ΙΠ-G), (III-H), (IVAA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IVBB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as set forth above, and any spécifie substituent set forth herein for a R1, Ra, X, W, Y1, Y2, L, Z1, Z2, Z3, or Z4 group in the compounds of Formulas (I), (II-A), (II-B), (II-C), (III-A), (III-B), (III-C), (III-D), (III-E), (III-F), (ΙΠ-G), (ΙΠ-Η), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), as set forth above, may be independently combined with other embodiments and/or substituents of compounds of Formulas (I), (Π-A), (II-B), (II-C), (III-A), (ΠΙ-Β), (III-C), (ΙΠ-D), (ΠΙ-Ε), (ΠΙ-F), (ΠΙ-G), (ΙΠ-Η), (IV-AA), (IV-AB), (IVAC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IVBD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), to form embodiments of the inventions not specifîcally set forth above. In addition, in the event that a list of substitutents is listed for any particular R1, Ra, X, W, Y1, Y2, L, Z1, Z2, Z3, or Z4 in a particular embodiment and/or claim, it is understood that each individual substituent may be deleted from the particular embodment and/or claim and that the remaining list of substituents will be considered to be within the scope of the invention.
As one of skill in the art will appreciate, compounds of Formulas (I), (II-A), (II-B), (II-C), (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG), (IV-AH), (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG), and (IV-BH), wherein Z1 and Z2 or Z1 and Z3, taken together, form -L- may be shown in several different ways. For example, the Compound 3 of Example 3 may be shown as:
Pharmaceutical Compositions
For the purposes of administration, in certain embodiments, the compounds described herein are administered as a raw chemical or are formulated as pharmaceutical compositions. Pharmaceutical compositions disclosed herein include a compound of Formula (I) and one or more of: a pharmaceutically acceptable carrier, diluent or excipient. The compound of Formula (I) is présent in the composition in an amount which is effective to treat a particular disease or condition of interest. The activity of compounds of Formula (I) can be determined by one skilled in the art, for example, as described in the Examples below. Appropriate concentrations and dosages can be readily determined by one skilled in the art. In certain embodiments, a compound of Formula (I) is présent in the pharmaceutical composition in an amount from about 25 mg to about 500 mg. In certain embodiments, a compound of Formula (I) is présent in the pharmaceutical composition in an amount of about 100 mg to about 300 mg. In certain embodiments, a compound of Formula (I) is présent in the pharmaceutical composition in an amount of about 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg or about 500 mg.
Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, is carried out via any of the accepted modes of administration of agents for serving similar utilities. The pharmaceutical compositions of the invention are prepared by combining a compound of the invention with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and in spécifie embodiments are formulated into préparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aérosols. Exemplary routes of administering such pharmaceutical compositions include, without limitation, oral, topical, transdermal, inhalation, parentéral, sublingual, buccal, rectal, vaginal, and intranasal. Pharmaceutical compositions of the invention are formulated so as to allow the active ingrédients contained therein to be bioavailable upon administration of the composition to a patient. Compositions that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the invention in aérosol form may hold a plurality of dosage units. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 20th Edition (Philadelphia College of Pharmacy and Science, 2000). The composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable sait thereof, for treatment of a disease or condition of interest in accordance with the teachings described herein.
The pharmaceutical compositions disclosed herein are prepared by méthodologies well known in the pharmaceutical art. For example, in certain embodiments, a pharmaceutical composition intended to be administered by injection is prepared by combining a compound of the invention with stérile, distilled water so as to form a solution. In some embodiments, a surfactant is added to facilitate the formation of a homogeneous solution or suspension. Surfactants are compounds that non-covalently interact with the compound of the invention so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.
The compounds of the invention, or their pharmaceutically acceptable salts, are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the spécifie compound employed; the metabolic stability and length of action of the compound; the âge, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excrétion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.
Combination Therapv
In one embodiment, a method for treating or preventing an HIV infection in a human having or at risk of having the infection is provided, comprising administering 5 to the human a therapeutically effective amount of a compound disclosed herein, or a pharmaceutically acceptable sait thereof, in combination with a therapeutically effective amount of one or more additional therapeutic agents.
In one embodiment, pharmaceutical compositions comprising a compound disclosed herein, or a pharmaceutically acceptable sait thereof, in combination 10 with one or more additional therapeutic agents, and a pharmaceutically acceptable carrier, diluent or excipient are provided.
In one embodiment, combination pharmaceutical agents comprising a compound disclosed herein, or a pharmaceutically acceptable sait thereof, in combination with one or more additional therapeutic agents are provided.
In the above embodiments, the additional therapeutic agent may be an anti-HIV agent. For example, in some embodiments, the additional therapeutic agent is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucléotide inhibitors of reverse transcriptase, HTV integrase inhibitors, HIV non20 catalytic site (or allosteric) integrase inhibitors, entry inhibitors (e.g., CCR5 inhibitors, gp41 inhibitors (i.e., fusion inhibitors) and CD4 attachment inhibitors), CXCR4 inhibitors, gpl20 inhibitors, G6PD andNADH-oxidase inhibitors, compounds that target the HIV capsid (“capsid inhibitors”; e.g., capsid polymerization inhibitors or capsid disrupting compounds such as those disclosed in WO 2013/006738 (Gilead Sciences),
US 2013/0165489 (University of Pennsylvania), and WO 2013/006792 (Pharma Resources), pharmacokinetic enhancers, and other drugs for treating HIV, and combinations thereof In further embodiments, the additional therapeutic agent is selected from one or more of:
(1) HIV protease inhibitors selected from the group consisting of amprenavir, atazanavir, fosamprenavir, indinavir, lopinavir, ritonavir, nelfmavir, saquinavir, tipranavir, brecanavir, darunavir, TMC-126, TMC-114, mozenavir (DMP
450), JE-2147 (AGI 776), L-756423, RO0334649, KNI-272, DPC-681, DPC-684, GW640385X, DG17, PPL-100, DG35, and AG 1859;
(2) HIV non-nucleoside or non-nucleotide inhibitors of reverse transcriptase selected from the group consisting of capravirine, emivirine, delaviridine, efavirenz, nevirapine, (+) calanolide A, etravirine, GW5634, DPC-083, DPC-961, DPC963, MIV-150, TMC-120, rilpivirene, BILR 355 BS, VRX 840773, lersivirine (UK453061), RDEA806, KM023 and MK-1439;
(3) HIV nucleoside inhibitors of reverse transcriptase selected from the group consisting of zidovudine, emtricitabine, didanosine, stavudine, zalcitabine, lamivudine, abacavir, amdoxovir, elvucitabine, alovudine, MIV-210, ±-FTC, D-d4FC, emtricitabine, phosphazide, fozivudine tidoxil, apricitibine (AVX754), KP-1461, GS9131 (Gilead Sciences) and fosalvudine tidoxil (formerly HDP 99.0003);
(4) HIV nucléotide inhibitors of reverse transcriptase selected from the group consisting of tenofovir, tenofovir disoproxil fumarate, tenofovir alafenamide fumarate (Gilead Sciences), GS-7340 (Gilead Sciences), GS-9148 (Gilead Sciences), adefovir, adefovir dipivoxil, CMX-001 (Chimerix) or CMX-157 (Chimerix);
(5) HIV integrase inhibitors selected from the group consisting of curcumin, dérivatives of curcumin, chicoric acid, dérivatives of chicoric acid, 3,5dicaffeoylquinic acid, dérivatives of 3,5-dicaffeoylquinic acid, aurintricarboxylic acid, dérivatives of aurintricarboxylic acid, caffeic acid phenethyl ester, dérivatives of caffeic acid phenethyl ester, tyrphostin, dérivatives of tyrphostin, quercetin, dérivatives of quercetin, S-1360, AR-177, L-870812, and L-870810, raltegravir, BMS-538158, GSK364735C, BMS-707035, MK-2048, BA 011, elvitegravir, dolutegravir and GSK744;
(6) HIV non-catalytic site, or allosteric, integrase inhibitors (NCINI) including, but not limited to, BI-224436, CX0516, CX05045, CX14442, compounds disclosed in WO 2009/062285 (Boehringer Ingelheim), WO 2010/130034 (Boehringer Ingelheim), WO 2013/159064 (Gilead Sciences), WO 2012/145728 (Gilead Sciences), WO 2012/003497 (Gilead Sciences), WO 2012/003498 (Gilead Sciences) each of which is incorporated by référencés in its entirety herein;
(7) gp41 inhibitors selected from the group consisting of enfuvirtide, sifuvirtide, albuvirtide, FB006M, and TRI-1144;
(8) the CXCR4 inhibitor AMD-070;
(9) the entry inhibitor SPOl A;
(10) the gpl20 inhibitor BMS-488043;
(11) the G6PD and NADH-oxidase inhibitor immunitin;
(12) CCR5 inhibitors selected from the group consisting of aplaviroc, vicriviroc, maraviroc, cenicriviroc, PRO-140, INCB15050, PF-232798 (Pfizer), and CCR5mAbOO4;
(13) CD4 attachment inhibitors selected from the group consisting of ibalizumab (TMB-355) and BMS-068 (BMS-663068);
(14) pharmacokinetic enhancers selected from the group consisting of cobicistat and SPI-452; and (15) other drugs for treating HIV selected from the group consisting of BAS-100, SPI-452, REP 9, SP-01 A, TNX-355, DES6, ODN-93, ODN-112, VGV-1, PA457 (bevirimat), HRG214, VGX-410, KD-247, AMZ 0026, CYT 99007A-221 HIV, DEBIO-025, BAY 50-4798, MDX010 (ipilimumab), PBS 119, ALG 889, and PA1050040 (PA-040), and combinations thereof
In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable sait thereof, is combined with two, three, four or more additional therapeutic agents. In certain embodiments, a compound disclosed herein, or a pharmaceutically acceptable sait thereof, is combined with two additional therapeutic agents. In other embodiments, a compound disclosed herein, or a pharmaceutically acceptable sait thereof, is combined with three additional therapeutic agents. In further embodiments, a compound disclosed herein, or a pharmaceutically acceptable sait thereof, is combined with four additional therapeutic agents. The two, three four or more additional therapeutic agents can be different therapeutic agents selected from the same class of therapeutic agents, or they can be selected from different classes of therapeutic agents. In a spécifie embodiment, a compound disclosed herein, or a pharmaceutically acceptable sait thereof, is combined with an HIV nucléotide inhibitor of reverse transcriptase and an HIV non-nucleoside inhibitor of reverse transcriptase. In another spécifie embodiment, a compound disclosed herein, or a pharmaceutically acceptable sait thereof, is combined with an HIV nucléotide inhibitor of reverse transcriptase, and an
HIV protease inhibiting compound. In a further embodiment, a compound disclosed herein, or a pharmaceutically acceptable sait thereof, is combined with an HIV nucléotide inhibitor of reverse transcriptase, an HIV non-nucleoside inhibitor of reverse transcriptase, and an HIV protease inhibiting compound. In an additional embodiment, a compound disclosed herein, or a pharmaceutically acceptable sait thereof, is combined with an HIV nucléotide inhibitor of reverse transcriptase, an HIV non-nucleoside inhibitor of reverse transcriptase, and a pharmacokinetic enhancer.
In certain embodiments, when a compound disclosed herein is combined with one or more additional therapeutic agents as described above, the components of the composition are administered as a simultaneous or sequential regimen. When administered sequentially, the combination may be administered in two or more administrations.
In certain embdoiments, a compound disclosed herein is combined with one or more additional therapeutic agents in a unitary dosage form for simultaneous administration to a patient, for example as a solid dosage form for oral administration.
In certain embodiments, a compound disclosed herein is administered with one or more additional therapeutic agents. Co-administration of a compound disclosed herein with one or more additional therapeutic agents generally refers to simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents, such that therapeutically effective amounts of the compound disclosed herein and one or more additional therapeutic agents are both présent in the body of the patient.
Co-administration includes administration of unit dosages of the compounds disclosed herein before or after administration of unit dosages of one or more additional therapeutic agents, for example, administration of the compound disclosed herein within seconds, minutes, or hours of the administration of one or more additional therapeutic agents. For example, in some embodiments, a unit dose of a compound disclosed herein is administered first, followed within seconds or minutes by administration of a unit dose of one or more additional therapeutic agents. Alternatively, in other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound disclosed herein within seconds or minutes. In some embodiments, a unit dose of a compound disclosed herein is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of one or more additional therapeutic agents. In other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed, after a period of hours (e.g., 1-12 hours), by administration of a unit dose of a compound disclosed herein.
The following Examples illustrate various methods of making compounds of this invention, i.e., compound of Formula (I):
wherein R1, X, W, Y1, Y2, Z1, Z2, or Z4 are as defined above. It is understood that one skilled in the art may be able to make these compounds by similar methods or by combining other methods known to one skilled in the art. It is also understood that one skilled in the art would be able to make, in a similar manner as described below, other compounds of Formula (I) not specifically illustrated below by using the appropriate starting components and modifying the parameters of the synthesis as needed. In general, starting components may be obtained from sources such as Sigma Aldrich, Lancaster Synthesis, Inc., Maybridge, Matrix Scientific, TCI, and Fluorochem USA, etc. or synthesized according to sources known to those skilled in the art (see, for example, Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5th édition (Wiley, December 2000)) or prepared as described herein.
The following examples are provided for purposes of illustration, not limitation.
EXAMPLES
General Synthetic Schemes
Schemes 1-3 are provided as further embodiments of the invention and illustrate general methods which were used to préparé compounds having Formula (I) 5 and which can be used to préparé additional compound having Formula (I).
Scheme 1
o O..
A1
R-°y° R o y, y2 r'°Yi 0H
HO
o o
A3
A5
A1
A2
A2
o Ox νΎ
H
A3
A1
A6
A8
A4
Al can be converted to amide A2 with an appropriate amine and a coupling reagent such as HATU or EDCI. A2 can be converted to A3 with a strong acid such as methanesulfonic acid. A3 can be converted to either A5 or A4 by heating with an appropriate cyclic diamine or cyclic aminoalcohol followed by methyl deprotection with 15 a reagent such as magnésium bromide.
Altematively, Al can be converted to A6 by treatment with a strong acid such as methanesulfonic acid. A6 can be condensed with an appropriate cyclic diamine or cyclic aminoalcohol followed by methyl deprotection with a reagent such as magnésium bromide to form either A7 or A8 respectively. A7 or A8 can be converted into amides A5 and A4 by treatment with an appropriate amine and a coupling reagent such as HATU or EDCI followed by methyl deprotection with a reagent such as 5 magnésium bromide.
Scheme 2
o OBn
O OBn
B1
B2
B1 (as described in W02012/018065) is condensed with diamine under reflux condition to give B2. B2 is hydrolyzed and coupled with an amine by an amide10 forming method to afford product B3 upon removal of a benzyl protecting group.
REPRESENTATIVE COMPOUNDS
Example 1
Préparation of Compound 1
N-(2,4-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13a-octahydro-2,515 methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
(+/-)
(+/-)
Step 1
-(2,2-dimethoxyethyl)-5-methoxy-6-(methoxycarbonyl)-4-oxo-1,4dihydropyridine-3-carboxylic acid (1-A, 0.300 g, 0.95 mmol), prepared as described in WO2011/119566 Al, was evaporated once from dry toluene, suspended in acetonitrile (4 mL) and treated with Ν,Ν-diisopropylethylamine (DIPEA) (0.329 mL, 1.90 mmol),
2,4-difluorobenzylamine (0.125 mL, 1.05 mmol) and HATU (0.433 g, 1.14 mmol). The reaction mixture was stirred for 10 minutes and concentrated. The residue was purified by flash chromatography on silica gel (10 to 60% ethyl acetate:dichloromethane) to afford the compound methyl 5-(2,4-difluorobenzylcarbamoyl)-l-(2,2-dimethoxyethyl)3-methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate, 1-B. 1H-NMR (400 MHz, DMSOd6) δ 10.28 (t, J= 6.0 Hz, IH), 8.46 (s, IH), 7.42 (dd, J = 15.4, 8.6 Hz, IH), 7.24 (m, IH), 7.06 (m, IH), 4.52 (m, 3H), 4.22 (d, J= 4.4 Hz, 2H), 3.92 (s, 3H), 3.80 (s, 3H),3.29 (d, 6H). LCMS-ESI+ (m/z)·. [M+H]+calculated for C20H23F2N2O7: 441.15; found: 441.2.
Step 2
Methyl 5-(2,4-difluorobenzylcarbamoyl)-1 -(2,2-dimethoxyethyl)-3methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-B, 0.106 g, 0.24 mmol) in acetonitrile (0.9 mL) and acetic acid (0.1 mL) was treated with methanesulfonic acid (0.005 mL, 0.072 mmol), sealed with a yellow cap, and heated to 70 °C. After 16 hours, the mixture was cooled to afford a crude solution of methyl 5-(2,4difluorobenzylcarbamoyl)-1 -(2,2-dihydroxyethyl)-3 -methoxy-4-oxo-1,4dihydropyridine-2-carboxylate, 1-C. LCMS-ESU (m/z)·. [M+H]+ calculated for C18H19F2N2O7: 413.12; found: 413.1.
Steps 3 and 4
Methyl 5-(2,4-difluorobenzylcarbamoyl)-l-(2,2-dihydroxyethyl)-3methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 0.65 mL of the crude mixture from the previous step, 0.17 mmol) was treated with acetonitrile (0.65 mL) and cis-3aminocyclpentanol (0.06 mL). The reaction mixture was sealed and heated to 90 °C. After 30 minutes, the reaction mixture was cooled and magnésium bromide (0.063 g, 0.34 mmol) was added. The mixture was resealed and heated to 50 °C. After 10 minutes, the reaction mixture was partitioned between dichloromethane and hydrochloric acid (0.2 M aq). The organic layer was removed and the aqueous layer extracted again with dichlormethane. The combined organic layers were dried over sodium sulfate, filtered and concentrated. Prep-HPLC purification (30-70% acetonitrile:water, 0.1% TFA) afforded Compound 1 as a racemic mixture. ’H-NMR (400 MHz, DMSO-d6/ δ 12.45 (br s, IH), 10.35 (t, J= 5.8 Hz, IH), 8.45 (s, IH), 7.37 (dd, J= 15.4, 8.6 Hz, IH), 7.23 (dt, J = 2.5, 9.9 Hz, IH), 7.05 (dt, J= 2.2, 8.7 Hz, IH), 5.43 (dd, J= 9.6, 4.0 Hz, IH), 5.09 (br s, IH), 4.68 (dd, J= 13.2, 4.0 Hz, IH), 4.59 (br s, IH), 4.53 (m, 2H), 4.02 (dd, J= 12.6, 9.4 Hz), 1.93 (br s, 4H), 1.83 (d, J= 12.0 Hz), 1.57 (dt, J= 12.2, 3.2 Hz). LCMS-ESI+ (m/z): [M+H]* calculated for C21H20F2N3O5: 432.14; found: 432.2.
Examples 2 and 3
Préparation of Compounds 2 and 3 (2R,5S,13aR)-N-(2,4-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide (2) and (2S,5R,13aS)-N-(2,4-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide (3)
Compound 1(16 mg) was separated by chiral HPLC using Chiralpak ASH with 100% éthanol as eluent to afford Compounds 2 and 3 in enantiomerically enriched form. For Compound 2: LCMS-ESU (m/z): [M+H]+ calculated for C21H20F2N3O5: 432.14; found: 432.2, Chiral HPLC rétention time = 4.50 minutes (Chiralpak AS-H, 150 5 x 4.6 mm, 1 mL/min EtOH). For Compound 3: LCMS-ESI+ (m/z): [M+H]calculated for C21H20F2N3O5: 432.14; found: 432.2, Chiral HPLC rétention time = 6.84 minutes (Chiralpak AS-H, 150 x 4.6 mm, 1 mL/min EtOH). ’H-NMR (400 MHz, DMSO-d6) δ 12.45 (br s, IH), 10.35 (t, J= 5.8 Hz, IH), 8.44 (s, IH), 7.37 (dd, J= 15.2, 8.4 Hz, IH), 7.23 (m, IH), 7.05 (dt, J= 1.8 Hz, 8.7 Hz, IH), 5.44 (dd, 9.6, 4.0 Hz), 5.09 (br s, IH), 10 4.68 (dd, 12.8, 4.0 Hz, IH), 4.59 (br s, IH), 4.53 (m, 2H), 4.02 (dd, J= 12.6, 9.4 Hz,
IH), 1.93 (br s, 4H), 1.83 (d, 12.4 Hz, IH), 1.57 (m, IH).
Altematively, Compound 3 was prepared as follows:
Chiral SFC
AD-H column
Methyl 5-(2,4-difluorobenzylcarbamoyl)-l-(2,2-dihydroxyethyl)-3methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 1.2 mmol in 5 mL of 9:1 acetonitrile: acetic acid containing 0.026 mL methanesulfonic acid) was treated with acetonitrile (5.0 mL) and cA-3-aminocyclpentanol (0.24 g, 2.4 mmol). The reaction mixture was sealed and heated to 90 °C. After 30 minutes, the reaction mixture was cooled, treated with potassium carbonate (0.332 g, 2.4 mmol), sealed and reheated to 90 °C. After 15 minutes, the mixture was cooled and partitioned between dichlormethane and hydrochloric acid (0.2 M aqueous). The organic layer was removed and the aqueous 10 solution was extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate (anhydrous), filtered and concentrated. The residue was purified by flash chromatography (0-8% éthanol (containing 11% saturated aqueous ammonium hydroxide) in dichloromethane) to afford Intermediate 1-D. LCMS-ESI+ (m/z)·. [M+H]+calculated for C22H22F2N3O5: 446.15; found: 446.2
Intermediate 1-D (270 mg) was separated by chiral SFC on a 50 mm Chiralpak AD-H column using 50% (1:1 methanohacetonitrile) in supercritical carbon dioxide as eluent to afford Intermediates 3-A (first eluting peak) and 3-B (second eluting peak) in enantioenriched form. For 3-A: LCMS-ESI+ (m/z): [M+H]+ calculated for C22H22F2N3O5: 446.15; found: 446.2. For 3-B: LCMS-ESI+ (m/z): [M+H]+ calculated for C22H22F2N3O5: 446.15; found: 446.2.
Intermediate 3-A (0.110 g, 0.247 mmol) in acetonitrile (5 mL) was treated portion wise with magnésium bromide (0.091 g, 0.494 mmol), sealed and heated to 50 °C. After 10 minutes the mixture was cooled and partitioned between dichloromethane and hydrochloric acid (0.2 M aqueous). The organic layer was separated and the aqueous extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate, filtered and concentrated. Préparative HPLC purification (30-70% acetonitrile:water, 0.1% TFA) afforded Compound 3 in enantioenriched form. Chiral HPLC rétention time = 6.51 minutes (Chiralpak AS-H, 150 x 4.6 mm, 1 mL/min EtOH). LCMS-ESI+ (m/z): [M+H]+calculated for C21H20F2N3O5: 432.14; found: 432.2. *HNMR (400 MHz, DMSO-d6) δ 12.45 (br s, IH), 10.35 (t, J= 5.8 Hz, IH), 8.44 (s, IH), 7.37 (dd, J= 15.2, 8.4 Hz, IH), 7.23 (m, IH), 7.05 (dt, J= 1.8 Hz, 8.7 Hz, IH), 5.44 (dd, J= 9.6, 4.0 Hz), 5.09 (br s, IH), 4.68 (dd, J= 12.8, 4.0 Hz, IH), 4.59 (br s, IH), 4.53 (m, 2H), 4.02 (dd, J= 12.6, 9.4 Hz, IH), 1.93 (br s, 4H), 1.83 (d, J= 12.4 Hz, IH), 1.57 (m, IH).
Example 4
Préparation of Compound 4 (1 S,4R)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H-
1,4-methanopyrido[l ',2':4,5]pyrazino[ 1,2-a]pyrimidine-9-carboxamide
Methyl 5-(2,4-difluorobenzylcarbamoyl)-1-(2,2-dihydroxyethy 1)-3- methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 0.12 mmol in 0.53 mL of 9:1 acetonitrile:acetic acid containing 0.002 mL methanesulfonic acid) was treated with acetonitrile then (R)-pyrrolidin-3-amine (0.032 mL, 0.36 mmol). The reaction mixture was capped and heated to 90 °C for 5.5 hours. After cooling, the mixture was partitioned between dichloromethane and sodium bicarbonate (IM aqueous). The organic layer was separated and the aqueous was extracted again with ethyl acetate. The combined organic layers were dried over sodium sulfate (anhydrous), filtered and concentrated. The residue was dissolved in acetonitrile (1 mL), treated with magnésium bromide (0.022 g, 0.12 mmol), capped and heated to 50 °C for 10 minutes. After cooling the mixture was partitioned between dichloromethane and ammonium chloride (sat). The organic layer was separated and the aqueous was extracted again with dichloromethane. The aqueous layer was adjusted to pH = 1 with HCl (aq) and extracted again with dichloromethane. The aqueous solution was adjusted to pH = 3 with NaOH (aq) and extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate, filtered, and concentrated. Préparative HPLC purification (10-55% acetonitrile:water, 0.1% TFA) afforded Compound 4. ^-NMR (400 MHz, CD3OD-d4/ Ô 8.42 (s, IH), 7.42, (q, J= 7.7 Hz, IH), 6.99 - 6.90 (m, 2H), 5.07 (br s, IH), 4.73 (br d, 10.8 Hz, IH), 4.62 (s, 2H), 4.51 (br d, J= 12.8 Hz, IH), 4.07 (t, J= 11.8 Hz, IH), 3.4- 3.0 (m, 3H), 2.76 (br d, J = 8.8 Hz, IH), 2.15-2.0 (m, IH), 1.9-1.8 (m, IH). LCMS-ESH (m/z): [M+H]+calculated for C20H19F2N4O4: 417.14; found: 417.2.
Example 5
Préparation of Compound 5 (4R, 12aS)-N-(l-(2,4-difluorophenyl)cyclopropyl)-7-hydroxy-4-methyl-6,8-dioxo3,4,6,8,12,12a-hexahydro-2H-[l,3]oxazino[3,2-d]pyrido[l,2-a]pyrazine-9-carboxamide
5-B 'Ό Ο
5-C
Step 1 (4R, 12aS)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro2H-[l,3]oxazino[3,2-d]pyrido[l,2-a]pyrazine-9-carboxylic acid (Intermediate 5-A) was prepared in an analogous manner to (3S,llaR)-6-methoxy-3-methyl-5,7-dioxo2,3,5,7,11,1 la-hexahydrooxazolo[3,2-d]pyrido[l,2-a]pyrazine-8-carboxylic acid as described in WO2011/119566, substituting (R)-3-aminobutan-l-ol for (S)-2aminopropan-l-ol. WO2011/119566 is incorporated herein by reference in its entirety. A suspension of Intermediate 5-A (24.8 mg, 0.080 mmol), 1-(2,4difluorophenyl)cyclopropanamine HCl sait (5-B, 21.9 mg, 0.107 mmol), and HATU (48 mg, 0.126 mmol) in CH2Q2 (2 mL) was stirred at ambient température as N,Ndiisopropylethylamine (DIPEA) (0.1 mL, 0.574 mmol) was added. After 30 minutes, the reaction mixture was diluted with ethyl acetate before washing with 10% aqueous citric acid solution (xl) and saturated aqueous NaHCCL solution (xl). After the aqueous fractions were extracted with ethyl acetate (xl), the organic fractions were combined, dried (MgSCU), and concentrated. The residue was purified by combiflash (12 g column) using hexanes, ethyl acetate, and 20% methanol in ethyl acetate to obtain (4R,12aS)-N( 1-(2,4-difluorophenyl)cyclopropyl)-7-methoxy-4-methyl-6,8-dioxo-3,4,6,8,12,12ahexahydro-2H-[l,3]oxazino[3,2-d]pyrido[l,2-a]pyrazine-9-carboxamide, Intermediate 5-C. LCMS-ESI+ (m/z): [M+H]+ calculated for C23H24F2N3O5: 460.17; found 460.2.
Step 2
A suspension of Intermediate 5-C (39 mg, 0.080 mmol) and magnésium bromide (42 mg, 0.2282 mmol) in acetonitrile (2 mL) was stirred at 50 °C. After 1 hour, the reaction mixture was stirred at 0 °C bath when 1 N HCl (2 mL) was added. After the resulting mixture was diluted with water (~20 mL), the product was extracted with dichloromethane (x3) and the combined extracts were dried (MgSCL) and concentrated. The residue was purified by préparative HPLC to obtain (4R, 12aS)-N-( 1-(2,4difluorophenyl)cyclopropyl)-7-hydroxy-4-methyl-6,8-dioxo-3,4,6,8,12,12a-hexahydro2H-[l,3]oxazino[3,2-d]pyrido[l,2-a]pyrazine-9-carboxamide, compound 5, as TFA sait. ’H-NMR (400 MHz, CDChJ δ 10.72 (br s, IH), 8.37 (s, IH), 7.57 (d, J = 7.9 Hz, IH), 6.71-6.81 (m, 2H), 5.23 (dd, J = 5.6 and 4.4 Hz, IH), 4.98 (br quint, J = ~6.5 Hz, IH), 4.26 (dd, J = 13.6 and 4.4 Hz, IH), 4.12 (dd, J = 13.6 and 5.6 Hz, IH), 4.00-4.06 (m, 2H), 2.16-2.25 (m, IH), 1.55 (br dd, J = 13.8 and 1.8 Hz, IH), 1.40 (d, J = 6.8 Hz, 3H), 1.221.31 (m, 4H). 19F NMR (376.1 MHz, CDC13) δ -76.38 (s, 3F), -111.69 ~ -111.645 (m, 2F). LCMS-ESI+ (m/z): [M+H]+ calculated for CiîHzzFsNsOs: 446.15; found: 446.2.
Example 6
Préparation of Compound 6 (lR,4S)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-3,4,6,8,12,12a-hexahydro-2H- l,4-methanopyrido[T,2':4,5]pyrazino[l,2-a]pyrimidine-9-carboxamide
Methyl 5-(2,4-difluorobenzylcarbamoyl)-1 -(2,2-dihydroxyethyl)-3 methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 0.100 g, 0.243 mmol), (S)pyrrolidin-3-amine (0.043 mL, 0.485 mmol) and potassium carbonate (0.067 g, 0.485 mmol) were suspended in acetonitrile (1.9 mL) and acetic acid (0.1 mL) and heated to 90 °C for 1.5 hours. After cooling, the mixture was treated with magnésium bromide (0.090 g) and heated to 50 °C for 30 minutes. After cooling, the mixture partitioned between dichloromethane and 0.2 M HCl. The organic layer was separated and the aqueous was extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate (anhydrous), filtered and concentrated. Préparative HPLC purification (25-50% acetonitrile.water, 0.1% TFA) afforded Compound 6. ^-NMR (400 MHz, DMSO-ûfc) δ 10.33 (t, J= 6.0 Hz, IH), 8.44 (s, IH), 7.48 - 7.32 (m, IH), 7.31 -7.15(m, IH), 7.14-6.97 (m, IH), 4.86 (d, J = 2.9 Hz, IH), 4.62-4.54 (m, IH), 4.52 (d, ../=5.9 Hz, IH), 4.01 (d,J=13.0Hz, IH), 2.99-2.76 (m, 3H), 1.96-1.81 (m, IH), 1.71 - 1.53 (m, IH). LCMS-ESU (m/z): [M+H]+calculated for C20H19F2N4O4: 417.14; found: 417.2.
Example 7
Préparation of Compound 7 (2S,6R)-N-(2,4-difluorobenzyl)-9-hydroxy-8,10-dioxo-3,4,5,6,8,10,14,14a-octahydro2H-2,6-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazocine-ll-carboxamide
Methyl 5-(2,4-difluorobenzylcarbamoyl)-1 -(2,2-dihydroxyethyl)-3methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 0.050 g, 0.121 mmol), (1S,3R)3-aminocyclohexanol (0.028 g, 0.243 mmol) and potassium carbonate (0.034 g, 0.243 mmol) were suspended in acetonitrile (0.95 mL) and heated to 90 °C for 0.5 hour. After cooling, acetic acid (0.050 mL) was added and the mixture was reheated to 90 °C for 2h. After cooling the mixture was treated with magnésium bromide (0.044 g) and heated to 50 °C for 1 hour. After cooling, a second portion of magnésium bromide (0.044 g) was added and the mixture was reheated to 50 °C for 15 minutes. After cooling, the mixture partitioned between dichloromethane and 0.2 M HCl. The organic layer was separated and the aqueous was extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate (anhydrous), filtered and concentrated. Préparative HPLC purification (40-80% acetonitrile:water, 0.1% TFA) afforded Compound 7. 1H-NMR (400 MHz, DMSO-Æ) δ 12.40 (s, IH), 10.36 (t, 6.1 Hz, IH), 8.45 (s, IH), 7.48 - 7.29 (m, IH), 7.31 - 7.13 (m, IH), 7.13 - 6.97 (m, IH), 5.56 (dd, 10.0, 4.1 Hz, IH), 4.70 (dd, J = 12.7, 4.1 Hz, IH), 4.52 (d, J = 5.5 Hz, 2H), 4.40 - 4.29 (m, 2H), 4.06 (dd, J =
5, 10.2 Hz, IH), 2.46 - 2.36 (m, IH), 1.98 - 1.63 (m, 4H), 1.57 - 1.30 (m, 3H).
LCMS-ESI+ (m/z): [M+H]+calculated for C22H22F2N3O5: 446.15; found: 446.2.
Example 8
Préparation of Compound 8 (2R,6S)-N-(2,4-difluorobenzyl)-9-hydroxy-8,10-dioxo-3,4,5,6,8,10,14,14a-octahydro2H-2,6-methanopyrido[ 1 ',2':4,5]pyrazino[2,1 -b][l,3]oxazocine-11 -carboxamide
Compound 8 was prepared in a similar manner to compound 7 using (lR,3S)-3-aminocyclohexanol in place of (lS,3R)-3-aminocyclohexanol. 1H-NMR (400 MHz, DMSO-dô) Ô 12.40 (s, IH), 10.36 (t, J= 6.1 Hz, IH), 8.45 (s, IH), 7.48 - 7.30 (m, IH), 7.23 (td, J= 10.6, 2.7 Hz, IH), 7.05 (td, J= 8.3, 2.3 Hz, IH), 5.56 (dd, J= 10.1, 4.1 Hz, IH), 4.70 (dd, J= 12.8, 3.9 Hz, IH), 4.52 (d, J= 5.6 Hz, 2H), 4.39 - 4.27 (m, 2H), 4.06 (dd, J= 12.6, 10.0 Hz, IH), 2.47 - 2.35 (m, IH), 2.00 - 1.64 (m, 4H), 1.58 - 1.30 (m, 3H). LCMS-ESI+ (m/z)'. [M+H]+calculated for C22H22F2N3O5: 446.15; found: 446.2.
Examples 9 and 10
Préparation of Compounds 9 and 10 (2S,5R, 13aS)-N-((R)-l-(4-fluorophenyl)ethyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide and (2R,5S,13aR)-N-((R)-l-(4-fluorophenyl)ethyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb][l,3]oxazepine-10-carboxamide 10
O OH
Step 3
OH +/- °'5 nh2
Step 1 l-(2,2-dimethoxyethyl)-5-methoxy-6-(methoxycarbonyl)-4-oxo-l,4dihydropyridine-3-carboxylic acid (1-A, 0.500 g, 1.59 mmol), was suspended in 5 acetonitrile (6 mL) and treated with Ν,Ν-diisopropylethylamine (DIPEA) (0.550 mL,
3.17 mmol), (R)-l-(4-fluorophenyl)ethanamine (0.242 mg, 1.74 mmol) and HATU (0.661 g, 1.74 mmol). The reaction mixture was stirred for 2 hours and partitioned between ethyl acetate and water. The organic layer was separated and washed with HCl (10% aq), sodium bicarbonate (IM aq), dried over sodium sulfate, filtered and 10 concentrated to afford crude (R)-methyl l-(2,2-dimethoxyethyl)-5-(l-(4fluorophenyl)ethylcarbamoyl)-3 -methoxy-4-oxo-1,4-dihydropyridine-2-carboxylate which was used without purification in the next step: LCMS-ESI+ (m/zy [M+H]+ calculated for C21H26FN2O7: 437.17; found: 437.1.
Step 2 (R)-methyl l-(2,2-dimethoxyethyl)-5-(l-(4fluorophenyl)ethylcarbamoyl)-3 -methoxy-4-oxo-1,4-dihydropyridine-2-carboxylate was suspended in acetonitrile (5.7 mL) and acetic acid (0.6 mL) and treated with methane sulfonic acid (0.031 mL, 0.477 mmol). The mixture was capped and heated to 75 °C.
After 7h, the mixture was cooled and used without purification in the next step: LCMSESI+ (m/z): [M+H]+calculated for C19H22FN2O7: 409.14; found: 409.0.
Step 3 (R)-methy l 1 -(2,2-dihydroxyethy 1)-5-( 1 -(4fluorophenyl)ethylcarbamoyl)-3 -methoxy-4-oxo-1,4-dihydropyridine-2-carboxylate (3.6 mL of the crude mixture from Step 2, 0.8 mmol) was diluted with acetonitrile (3.6 mL) and treated with czs-3-aminocyclpentanol, HCl sait (0.219 g, 1.6 mmol) and potassium carbonate (0.276 g, 2.0 mmol). The mixture was capped and heated to 90 °C. After 20 minutes, the reaction mixture was cooled and partitioned between dichloromethane and HCl (0.2 M aq). The layers were separated and the aqueous layer was extracted again with dichloromethane. The combined organic layers were treated with a small amount of acetonitrile, dried over sodium sulfate, fîltered and concentrated.
The residue was suspended in acetonitrile (4 mL) and treated with magnésium bromide (0.177 g). The mixture was capped and heated to 50 °C. After 10 minutes, the reaction mixture was cooled and partitioned between dichloromethane and HCl (0.2 M aq). The layers were separated and the aqueous layer was extracted again with dichlormethane. The combined organic layers were dried over sodium sulfate, fîltered and concentrated. The residue was purified by flash chromatography on silica gel (0-8% ethanof.DCM) to afford a diastereomeric mixture of desired 9 and 10.
The mixture was separated by chiral HPLC using Chiralpak AD-H with 100% éthanol as eluent to afford Compounds 9 and 10 in enantiomerically enriched form:
For Compound 9: LCMS-ESI+ (m/z): [M+H]+ calculated for
C22H23FN3O5: 428.16; found: 428.1. Chiral HPLC rétention time = 10.177 minutes (Chiralpak AD-H, 150 x 4.6 mm, 1 mL/min EtOH). Ή-NMR (400 MHz, DMSO-de) δ 12.45 (s, IH), 10.45 (d, .7= 7.7 Hz, IH), 8.40 (s, IH), 7.37 (dd, J= 8.6, 5.6 Hz, 2H), 7.15 (t, J= 8.9 Hz, 2H), 5.44 (dd, J= 9.5, 4.2 Hz, IH), 5.17 - 5.04 (m, 2H), 4.73 - 4.62 (m, IH), 4.59 (s, IH), 4.00 (dd, J= 12.7, 9.5 Hz, IH), 1.93 (s, 4H), 1.83 (d, J= 11.8 Hz, IH), 1.56 (dt, J= 12.1, 3.4 Hz, IH), 1.44 (d, .7=6,9 Hz, 3H).
For Compound 10: LCMS-ESI+ (m/z): [M+H]+ calculated for C22H23FN3O5: 428.16; found: 428.1. Chiral HPLC rétention time = 14.061 minutes (Chiralpak AD-H, 150 x 4.6 mm, 1 mL/min EtOH). ‘H-NMR (400 MHz, DMSO-J6) δ
12.44 (s, IH), 10.46 (d, J= 7.8 Hz, IH), 8.41 (s, IH), 7.37 (dd, J= 8.6, 5.6 Hz, 2H), 7.15 (t, J= 8.9 Hz, 2H), 5.42 (dd, 9.6, 4.1 Hz, IH), 5.18- 5.02 (m, 2H), 4.67 (dd, J= 12.8, 4.2 Hz, IH), 4.59 (s, IH), 4.02 (dd, 12.7, 9.6 Hz, IH), 1.93 (s, 4H), 1.83 (d, J= 12.0 Hz, IH), 1.57 (dt, J= 13.0, 3.5 Hz, IH), 1.44 (d, J= 6.9 Hz, 3H).
Example 11
Préparation of Compound 11 (2S,5R,13aS)-N-((R)-l-(2,4-difluorophenyl)ethyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb] [ 1,3 ]oxazepine-10-carboxamide
'nh2
Step 1 l-(2,2-dimethoxyethyl)-5-methoxy-6-(methoxycarbonyl)-4-oxo-l,415 dihydropyridine-3-carboxylic acid (1-A, 0.315 g, 1.00 mmol), was suspended in acetonitrile (4 mL) and treated with Ν,Ν-diisopropylethylamine (DIPEA) (0.348 mL, 2.00 mmol), (R)-l-(2,4-difluorophenyl)ethanamine HCl sait (0.213 mg, 1.10 mmol) and HATU (0.418 g, 1.10 mmol). The reaction mixture was stirred for 1 hour and partitioned between dichloromethane and HCl (10% aq). The organic layer was separated and washed sodium bicarbonate (IM aq), dried over sodium sulfate, filtered and concentrated to afford crude (R)-methyl 5-(l-(2,4-difluorophenyl)ethylcarbamoyl)-l-(2,2dimethoxyethyl)-3-methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate which was used without purification in the next step. LCMS-ESI+ (m/z): [M+H]+ calculated for C21H25F2N2O7: 455.16; found: 455.1.
Step 2 (R)-methyl 5-(l-(2,4-difluorophenyl)ethylcarbamoyl)-l-(2,2dimethoxyethyl)-3-methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate was suspended in acetonitrile (3.6 mL) and acetic acid (0.4 mL) and treated with methane sulfonic acid (0.020 mL). The mixture was capped and heated to 75 °C. After 16 hours, the crude mixture was cooled and used without purification in the next step. LCMS-ESI+ (m/z): [M+H]+calculated for C19H21F2N2O7: 427.13; found: 427.1.
Step 3 (R)-methyl 5-( 1 -(2,4-difluorophenyl)ethylcarbamoyl)-1 -(2,2dihydroxyethyl)-3-methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (half of the crude mixture from Step 2, approx 0.5 mmol) was diluted with acetonitrile (2.5 mL) and treated with (lS,3R)-3-aminocyclopentanol (0.110 g, 1.09 mmol) and potassium carbonate (0.069 g, 0.50 mmol). The mixture was capped and heated to 90 °C. After 15 minutes, the reaction mixture was cooled and magnésium bromide (0.184 g) was added. The reaction mixture was heated to 50 °C. After 10 minutes, the mixture was cooled and treated with an additional portion of magnésium bromide (0.184 g). The reaction mixture was reheated to 50 °C and stirred for 10 minutes. After cooling, the mixture was partitioned between dichloromethane and HCl (0.2 M aq). The layers were separated and the aqueous layer was extracted again with dichloromethane. The combined organic layers were dried over sodium sulfate, filtered and concentrated. Préparative HPLC purification (30-60% acetonitrile:water, 0.1% TFA) afforded desired Compound 11. LCMS-ESI+ (m/z): [M+H]+calculated for C22H22F2N3O5: 446.15; found: 446.1. *11NMR (400 MHz, DMSO-de) δ 12.46 (s, IH), 10.53 (d, 7.5 Hz, IH), 8.38 (s, IH), 7.39 (q, J = 8.5 Hz, IH), 7.29 - 7.12 (m, IH), 7.13 - 6.93 (m, IH), 5.44 (dd, J = 9.8, 4.2 Hz, IH), 5.28 (p, 7.3, 6.8 Hz, IH), 5.09 (s, IH), 4.66 (dd, J= 13.2, 4.3 Hz, IH), 4.59 (s,
IH), 3.99 (dd, 7= 13.1, 9.6 Hz, IH), 1.93 (s, 4H), 1.83 (d, 7= 12.4 Hz, IH), 1.56 (dt, 7=
12.5, 2.9 Hz, IH), 1.45 (d, 7= 6.9 Hz, 3H).
Example 12
Préparation of Compound 12 (2R,5S,13aR)-N-((R)-l-(2,4-difluorophenyl)ethyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb][ 1,3]oxazepine-l 0-carboxamide
Compound 12 was prepared in a similar manner to compound 11 using (lR,3S)-3-aminocyclopentanol in place of (lS,3R)-3-aminocycIopentanoI. 1H-NMR (400 MHz, DMSO-76) Ô 12.43 (s, IH), 10.52 (d, 7= 8.2 Hz, IH), 8.38 (s, IH), 7.39 (q, 7 = 8.4 Hz, IH), 7.28 - 7.12 (m, IH), 7.11 - 6.97 (m, IH), 5.41 (dd, 7 = 10.0, 4.0 Hz, IH), 5.35 - 5.20 (m, IH), 5.08 (s, IH), 4.65 (dd, 7= 13.1, 3.8 Hz, IH), 4.58 (s, IH), 4.01 (dd, 7= 12.8, 9.5 Hz, IH), 1.92 (s, 4H), 1.83 (d, 7= 11.5 Hz, IH), 1.61 - 1.51 (m, IH), 1.44 (d, 7= 6.9 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+ calculaied for C22H22F2N3O5: 446.15; found: 446.1.
Example 13
Préparation of Compound 13 (2S,5R,13aS)-N-((S)-l-(2,4-difluorophenyl)ethyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[l',2':4,5]pyrazino[2,lb] [ 1,3 Joxazepine-10-carboxamide
Compound 13 was prepared in a similar manner to compound 11 using (S)-l-(2,4-difluorophenyl)ethanaminein place of (R)-l-(2,4-difluorophenyl)ethanamine, and using only a single portion of magnésium bromide (0.184 g). 1H-NMR (400 MHz, DMSO-tZô) δ 12.44 (s, IH), 10.53 (d, J= 7.8 Hz, IH), 8.39 (s, IH), 7.39 (q, J = 8.5 Hz, IH), 7.32 - 7.14 (m, IH), 7.05 (t, 9.1 Hz, IH), 5.42 (dd, J= 9.5, 4.2 Hz, IH), 5.29 (p,
6.9 Hz, IH), 5.09 (s, IH), 4.65 (dd, J= 12.9, 4.3 Hz, IH), 4.59 (s, IH), 4.02 (dd, J=
12.6, 9.8 Hz, IH), 1.92 (s, 4H), 1.83 (d, J= 12.1 Hz, IH), 1.61 - 1.52 (m, IH), 1.44 (d, J = 6.9 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+calculated for C22H22F2N3O5: 446.15; found:
446.2.
Example 14
Préparation of Compound 14 (2R,5S,13aR)-N-((S)-l-(2,4-difluorophenyl)ethyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb] [ 1,3 joxazepine-10-carboxamide
Compound 14 was prepared in a similar manner to compound 11 using (S)-l-(2,4-difluorophenyl)ethanamine in place of (R)-l-(2,4-difluorophenyl)ethanamine and using (lR,3S)-3-aminocyclopentanol in place of(lS,3R)-3-aminocyclopentanol. 1HNMR (400 MHz, DMSO-îZô) δ 12.46 (s, IH), 10.53 (d,J=7.6Hz, IH), 8.38 (s, IH), 7.39 (q, J= 8.6 Hz, IH), 7.28 - 7.14 (m, IH), 7.05 (t, J= 8.5 Hz, IH), 5.44 (dd, J= 9.8, 3.8 Hz, IH), 5.28 (p, J = 8.0 Hz, IH), 5.09 (s, IH), 4.66 (dd, J= 12.9, 4.0 Hz, IH), 4.59 (s,
IH), 3.99 (dd, J= 12.5, 9.6 Hz, IH), 1.93 (s, 4H), 1.83 (d, J= 12.6 Hz, IH), 1.56 (dt, J= 13.0, 3.3 Hz, IH), 1.45 (d, J = 6.9 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+calculated for C22H22F2N3O5: 446.15; found: 446.1.
Example 15
Préparation of Compound 15 (2S,5R,13aS)-N-(4-fluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13a-octahydro2,5-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Step 3
Step 1
-(2,2-dimethoxyethyl)-5-methoxy-6-(methoxycarbonyl)-4-oxo-1,4dihydropyridine-3-carboxylic acid (1-A, 3.15 g, 10.0 mmol), suspended in acetonitrile (36 mL) and acetic acid (4 mL) was treated with methane sulfonic acid (0.195 mL). The 15 mixture heated to 75 °C. After 7 hours, the crude mixture was cooled and stored in a 10°C for three days. The crude mixture was reheated to 75 °C for 2 hours, cooled used without purification in the next step. LCMS-ESI+ (m/zf. [M+H]+ calculated for C19H21F2N2O7: 288.07; found: 288.1.
Step 2
Crude l-(2,2-dihydroxyethyl)-5-methoxy-6-(methoxycarbonyl)-4-oxol,4-dihydropyridine-3-carboxylic acid (16.8 mL of crude mixture from Step 1, approx 4 mmol) was combined with (lS,3R)-3-aminocyclopentanol (0.809 g, 8 mmol), diluted with acetonitrile (16.8 mL), and treated with potassium carbonate (0.553 g, 4 mmol). The reaction mixture was heated to 85 °C, stirred for 15 minutes, cooled to ambient température and stirred an additional 16 hours. HCl (50 mL, 0.2M aq) was added and the clear yellow solution was extracted three times with dichloromethane. The combined organics were dried over sodium sulfate, filtered and concentrated to a yellow solid. This crude material was precipitated from dichloromethane/hexanes to afford desired intermediate 15-B as a light beige powder. 1H-NMR (400 MHz, DMSO-î/ô) δ 8.72 (s, IH), 5.42 (dd, J= 9.6, 4.1 Hz, IH), 5.09 (s, IH), 4.72 (dd, J= 13.0, 3.7 Hz, IH), 4.57 (s, IH), 4.09 (dd,/= 12.5, 9.6 Hz, IH), 3.83 (s, 3H), 1.92 (s, 3H), 1.78 (m, 2H), 1.62-1.47 (m, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for CisHnWDe: 321.11; found: 321.2.
Step 3
Intermediate 15-B (0.040 g, 0.125 mmol) and (4fluorophenyl)methanamine (0.017 g, 0.137 mmol) were suspended in acetonitrile (1 mL) and treated with Ν,Ν-diisopropylethylamine (DIPEA) (0.033 mL, 0.187 mmol) and HATU (0.052 g, 0.137 mmol). After stirring for 30 minutes, the reaction mixture was treated with magnésium bromide (0.046 g, 0.25 mmol) and heated to 50 °C. After 10 minutes, the reaction mixture was cooled and treated with HCl (2 mL, 10% aq). After a few minutes, the precipitate was filtered and washed with HCl (10% aq) and water. Préparative HPLC purification of the precipitate (20-65% acetonitrile:water, 0.1% TFA) afforded desired Compound 15. ’H-NMR (400 MHz, DMSO-dë) δ 12.44 (s, IH), 10.36 (t, J= 6.0 Hz, IH), 8.46 (s, IH), 7.37 - 7.28 (m, 2H), 7.19 - 7.09 (m, 2H), 5.43 (dd, J-
9.6, 4.0 Hz, IH), 5.08 (s, IH), 4.68 (dd, J= 12.8, 4.1 Hz, IH), 4.59 (s, IH), 4.58 - 4.42 (m, 3H), 4.02 (dd, J = 12.7, 9.6 Hz, IH), 1.92 (s, 5H), 1.83 (d, J= 12.2 Hz, IH), 1.56 (dt, J= 12.0, 3.4 Hz, IH). LCMS-ESI+ (m/z): [M+H]+calculated for C21H21FN3O5: 414.15; found: 414.2.
Example 16
Préparation of Compound 16 (2S,5R, 13aS)-N-(2,3-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5 -methanopyrido [ Γ, 2' : 4,5 ] pyrazino [2,1 -b] [ 1,3 ]oxazepine-10-carboxamide
Compound 16 was prepared in a similar manner to compound 15 using (2,3-difluorophenyl)methanamine in place of (4-fluorophenyl)methanamine. ^-NMR (400 MHz, DMSO-ùfc) δ 12.46 (s, IH), 10.41 (t, J= 6.1 Hz, IH), 8.45 (s, IH), 7.43 - 7.25 (m, IH), 7.25 - 7.05 (m, 2H), 5.44 (dd, ./ = 9.5, 3.9 Hz, IH), 5.09 (s, IH), 4.68 (dd, J = 12.8, 4.0 Hz, IH), 4.65-4.53 (m, 3H), 4.02 (dd, 12.7, 9.8 Hz, IH), 3.56 (s, IH), 1.93 (s, 4H), 1.83 (d, J= 11.9 Hz, IH), 1.57 (dt, J = 11.5, 3.0 Hz, IH). LCMS-ESF (m/z): [M+H]+calculated for C21H20F2N3O5: 432.14; found: 432.2.
Example 17
Préparation of Compound 17 (2S,5R, 13aS)-N-(4-chloro-2-fluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Compound 17 was prepared in a similar manner to compound 15 using (4chloro-2-fluorophenyl)methanamine in place of (4-fluorophenyl)methanamine. *ΗNMR (400 MHz, DMSO4) δ 12.46 (s, IH), 10.45 - 10.29 (m, IH), 8.44 (s, IH), 7.42 (dd, 10.0, 2.0 Hz, IH), 7.33 (t, J=8.1 Hz, IH), 7.26 (dd, J= 8.4, 1.8 Hz, IH), 5.50
5.38 (m, IH), 5.09 (s, IH), 4.68 (dd, J = 13.0, 4.0 Hz, IH), 4.59 (s, IH), 4.54 (m, 2H), 4.02 (dd,7=12.8, 9.7 Hz, IH), 1.93 (s, 4H), 1.83 (d, J= 12.0 Hz, IH), 1.57 (dt, 7= 11.9, 3.4 Hz, IH). LCMS-ESI+ (m/z); [M+H]+ calculated for C21H20CIFN3O5: 448.11; found:
448.2.
Example 18
Préparation of Compound 18 (2S,5R, 13aS)-N-(3,4-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[ 1 ',2':4,5]pyrazino[2,1 -b] [ 1,3 joxazepine-10-carboxamide
Compound 18 was prepared in a similar manner to compound 15 using (3,4-difluorophenyl)methanamine in place of (4-fluorophenyl)methanamine. 1H-NMR (400 MHz, DMSO-tie) δ 12.46 (s, IH), 10.51 - 10.27 (m, IH), 8.46 (s, IH), 7.50 - 7.23 (m, 2H), 7.23 - 7.03 (m, IH), 5.44 (dd, 7= 9.5, 3.6 Hz, IH), 5.09 (s, IH), 4.75 - 4.63 (m, IH), 4.60 (s, IH), 4.57-4.44 (m, 2H), 4.02 (dd, 7= 12.6, 9.8 Hz, IH), 1.93 (s, 4H), 1.83 (d, 7= 12.0 Hz, IH), 1.57 (dt, 7= 12.0, 3.4 Hz, IH). LCMS-ESI+ (m/z); [M+H]+ calculated for C21H20F2N3O5: 432.14; found: 432.2.
Example 19
Préparation of Compound 19 ( 1 R, 5 S)-N-(2,4-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13 a-octahydro-1,5methanopyrido[r,2':4,5]pyrazino[l,2-a][l,3]diazepine-10-carboxamide
Steps 1 and 2
Methyl 5-(2,4-difluorobenzylcarbamoyl)-l-(2,2-dihydroxyethyl)-3methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 97.5 mg, 0.236 mmol) was treated with acetonitrile (1.9 mL), acetic acid (0.1 mL), potassium carbonate (145 mg, 1.05 mmol), and (S)-piperidin-3-amine dihydrochloride (82 mg, 0.472 mmol). The reaction mixture was sealed and heated to 90 °C. After 60 minutes, the reaction mixture was cooled partitioned between brine and dichloromethane. The aqueous phase was thrice extracted into dichloromethane and the combined organic phases were combined, dried over MgSO4, fdtered, concentrated. The crude product was dissolved into acetonitrile (2 mL) and magnésium bromide (89.1 mg, 0.48 mmol) was added. The mixture was resealed and heated to 50 °C. After 90 minutes, the reaction mixture was quenched with ~5 mL of 0.2M HCl(aq), the pH adjusted to ~10, diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile:water, 0.1% TFA) afforded Compound 19. 1H-NMR (400 MHz, Chloroform-d) 5 10.43 (t, J = 5.9 Hz, IH), 8.43 (s, IH), 7.39 - 7.30 (m, IH), 6.81 (q, J = 8.1 Hz, 2H), 4.89 (dd, J = 11.6, 3.8 Hz, IH), 4.69 (s, IH), 4.64 (d, J = 5.8 Hz, 2H), 4.26 (dd, J = 12.6, 3.8 Hz, IH), 3.91 (t, J = 12.1 Hz, IH), 3.20 - 3.10 (m, 2H), 3.06 (s, 2H), 2.14 - 2.02 (m, IH), 1.96 - 1.81 (m, 2H), 1.81 1.70 (m, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H20F2N4O4: 431.15; found:
431.2.
Example 20
Préparation of Compound 20 (lS,5R)-N-(2,4-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13a-octahydro-l,5methanopyrido[r,2':4,5]pyrazino[l,2-a][l,3]diazepine-10-carboxamide
Steps 1 and 2
Methyl 5-(2,4-difluorobenzylcarbamoyl)-1 -(2,2-dihydroxyethyl)-3 methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 103.3 mg, 0.25 mmol) was treated with acetonitrile (1.9 mL), acetic acid (0.1 mL), potassium carbonate (159.8 mg,
I. 16 mmol), and (R)-piperidin-3-amine dihydrochloride (90 mg, 0.52 mmol). The reaction mixture was sealed and heated to 90 °C. After 40 minutes, the reaction mixture was cooled partitioned between brine and dichloromethane. The aqueous phase was thrice extracted into dichloromethane and the combined organic phases were combined, dried over MgSCL, filtered, concentrated. The crude product was dissolved into acetonitrile (2 mL) and magnésium bromide (96.5 mg, 0.52 mmol) was added. The mixture was resealed and heated to 50 °C. After 80 minutes, the reaction mixture was quenched with ~5 mL of 0.2M HCl (aq), the pH adjusted to ~10, diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile:water, 0.1% TFA) afforded Compound 20. Î-NMR (400 MHz, DMSO-de) δ 10.35 (t, J = 6.0 Hz, IH), 8.48 (s, IH), 7.45 - 7.33 (m, IH), 7.29 - 7.18 (m, IH), 7.05 (td, J = 8.5, 2.4 Hz, IH), 5.06 (dd, J =
II. 4, 3.5 Hz, IH), 4.56 - 4.47 (m, 3H), 4.44 (s, IH), 4.05 (t, J = 11.8 Hz, IH), 3.07 - 2.89 (m, 4H), 1.85 - 1.73 (m, 3H), 1.54-1.46 (m, IH). LCMS-ESI+ (m/z): [M+H]+calculated for C21H20F2N4O4: 431.15; found: 431.2.
Example 21
Préparation of Compound 21 (2 S, 5R, 13 aS)-N-((S)-1 -(4-fluorophenyl)ethyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13 aoctahydro-2,5-methanopyrido[T,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Steps 1 and 2 (S)-Methyl l-(2,2-dihydroxyethyl)-5-(l-(4fluorophenyl)ethylcarbamoyl)-3-methoxy-4-oxo-1,4-dihydiOpyridine-2-carboxylate (21-A, 1 mL, 0.23 M solution in 19.1 acetonitrile:acetic acid, prepared as per (R)-methyl l-(2,2-dihydroxyethyl)-5-(l-(4-fluorophenyl)ethylcarbamoyl)-3-methoxy-4-oxo-l,4dihydropyridine-2-carboxylate 9-A from Example 9 using (S)-l-(4fluorophenyl)ethanamine in place of (R)-l-(4-fluorophenyl)ethanamine) was treated with (lS,3R)-3-aminocyclopentanol (62 mg, 0.61 mmol) and potassium carbonate (34 mg, 0.25 mmol). The reaction mixture was sealed and heated to 90 °C. After 60 minutes, the reaction mixture was cooled partitioned between brine and dichloromethane. The aqueous phase was thrice extracted into dichloromethane and the combined organic phases were combined, dried over MgSCU, filtered, and concentrated. The crude product was dissolved into acetonitrile (2 mL) and magnésium bromide (74 mg, 0.4 mmol) was added. The mixture was resealed and heated to 50 °C. After 100 minutes, the reaction mixture was quenched with 0.2M HCl (aq), diluted with brine, and thrice extracted into DCM. HPLC purification (acetonitrile:water, 0.1% TFA) afforded Compound 21. *HNMR (400 MHz, DMSO-de) δ 12.42 (br s, IH), 10.45 (d, J = 7.9 Hz, IH), 8.40 (s, IH), 7.36 (dd, J = 8.6, 5.5 Hz, 2H), 7.14 (t, J = 8.9 Hz, 2H), 5.42 (dd, J = 9.6, 4.2 Hz, IH),
5.15 - 5.04 (m, 2Η), 4.72 - 4.55 (m, 2H), 4.02 (dd, J = 12.7, 9.7 Hz, 1Η), 1.97 - 1.89 (m, 4Η), 1.82 (d, J = 12.2 Hz, IH), 1.56 (dt, J = 11.9, 3.3 Hz, 1Η), 1.43 (d, J - 6.9 Hz, 3Η). LCMS-ESI+ (m/z): [M+H]+calculated for C22H22FN3O5: 428.16; found: 428.1.
Example 22
Préparation of Compound 22 (2R, 5 S, 13aR)-N-((S)-l-(4-fluorophenyl)ethyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13 aoctahydro-2,5-methanopyrido[ 1 ',2' : 4,5]pyrazino [2,1 -b] [ 1,3 Joxazepine-10-carboxamide
0 Â
21-A
Steps 1 and 2 (S)-methyl 1 -(2,2-dihydroxyethyl)-5-(l -(4fluorophenyl)ethylcarbamoyl)-3 -methoxy-4-oxo-1,4-dihydropyridine-2-carboxylate (21-A, 1 mL, 0.23 M solution in 19:1 acetonitrile:acetic acid) was treated with (1R,3S)3-aminocyclopentanol (52 mg, 0.51 mmol) and potassium carbonate (31 mg, 0.22 mmol). The reaction mixture was sealed and heated to 90 °C. After 60 minutes, the reaction mixture was cooled partitioned between brine and dichloromethane. The aqueous phase was thrice extracted into dichloromethane and the combined organic phases were combined, dried over MgSCL, filtered, and concentrated. The crude product was dissolved into acetonitrile (2 mL) and magnésium bromide (91 mg, 0.49 mmol) was added. The mixture was resealed and heated to 50 °C. After 100 minutes, the reaction mixture was quenched with 0.2M HCl(aq), diluted with brine, and thrice extracted into DCM. HPLC purification (acetonitrile.water, 0.1% TFA) afforded Compound 22. *H67
NMR (400 MHz, DMSO-d6) δ 12.44 (br s, IH), 10.45 (d, J = 7.7 Hz, IH), 8.39 (s, IH), 7.36 (dd, J = 8.5, 5.6 Hz, 2H), 7.14 (t, J = 8.9 Hz, 2H), 5.43 (dd, J = 9.6, 4.0 Hz, IH), 5.15-5.06 (m, 2H), 4.66 (dd, J= 12.8, 3.9 Hz, IH), 4.58 (s, IH), 3.99 (dd, J= 12.6, 9.5 Hz, IH), 1.93 (s, 4H), 1.82 (d, J = 12.0 Hz, IH), 1.56 (dt, J = 12.0, 3.0 Hz, IH), 1.44 (d, J = 6.9 Hz, 3H). LCMS-ESH (m/z): [M+H]+ calculated for C22H22FN3O5: 428.16; found: 428.1.
Example 23
Préparation of Compound 23 (2S,5R,13aS)-N-(2-fluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13a-octahydro2,5-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
O OH
Steps 1 and 2
15-B (41 mg, 0.13 mmol) was treated with acetonitrile (1 mL), (2fluorophenyl)methanamine (17 mg, 0.14 mmol), HATU (67 mg, 0.18 mmol), and N,Ndiisopropylethylamine (DIPEA) (24 mg, 0.19 mmol). The reaction mixture was stirred at room température for one hour and magnésium bromide (47 mg, 0.26 mmol) was added. The mixture was sealed and heated to 50 °C. After 60 minutes, the reaction mixture was quenched with 0.2M HCl (aq), diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile:water, 0.1% TFA) afforded Compound 23. 1H-NMR (400 MHz, Chloroform-d) δ 10.42 (s, IH), 8.34 (s, IH), 7.36 (t, J = 7.9 Hz, IH), 7.24 - 7.17 (m, IH), 7.12 - 6.97 (m, 2H), 5.40 - 5.32 (m, IH), 5.29 (t, J = 3.5 Hz, IH), 4.67 (s, 3H), 4.28-4.20 (m, IH), 4.06-3.95 (m, IH), 2.20- 1.96 (m, 4H), 1.95 - 1.84 (m, IH), 1.59 (dt, J = 12.4, 3.3 Hz, IH). LCMS-ESF (m/z): [M+H]+ calculated for C21H20FN3O5:
414.15; found: 414.2.
Example 24
Préparation of Compound 24 (2S,5R,13aS)-N-(3,5-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Steps 1 and 2 15-B (44 mg, 0.14 mmol) was treated with acetonitrile (1 mL), (3,5difluorophenyl)methanamine (32 mg, 0.23 mmol), HATU (54 mg, 0.14 mmol), and N,Ndiisopropylethylamine (37 mg, 0.29 mmol). The reaction mixture was stirred at room température for one hour and magnésium bromide (57 mg, 0.31 mmol) was added. The mixture was sealed and heated to 50 °C. After 60 minutes, the reaction mixture was quenched with 0.2M HCl (aq), diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile:water, 0.1% TFA) afforded Compound 24. 1H-NMR (400 5 MHz, Chloroform-d) δ 10.39 (s, IH), 8.42 (s, IH), 6.82 (d, J = 7.9 Hz, 2H), 6.65 (t, J =
8.8 Hz, IH), 5.38 (d, J = 7.7 Hz, 1 H), 5.28 (s, IH), 4.78-4.41 (m, 3H), 4.32 (d, J = 12.1 Hz, IH), 4.02 (t, J = 10.9 Hz, IH), 2.30- 1.97 (m, 4H), 1.97 - 1.81 (m, IH), 1.59 (d, J =
12.3 Hz, IH). LCMS-ESF (m/z): [M+H]+ calculated for C2iHi9F2N3O5: 432.14; found:
432.2.
Example 25
Préparation of Compound 25 (2S,5R,13aS)-N-(4-fluoro-3-(trifluoromethyl)benzyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13 a-octahydro-2,5 -methanopyrido [ l',2' : 4,5 ] pyrazino [2,115 b] [ 1,3 Joxazepine-10-carboxamide
Steps 1 and 2
15-B (43 mg, 0.13 mmol) was treated with acetonitrile (1 mL), (4-fluoro3-(trifluoromethyl)phenyl)methanamine (29 mg, 0.15 mmol), HATU (62 mg, 0.16 mmol), and N,N-diisopropylethylamine (26 mg, 0.20 mmol). The reaction mixture was stirred at room température for one hour and magnésium bromide (62 mg, 0.34 mmol) was added. The mixture was sealed and heated to 50 °C. After 60 minutes, the reaction mixture was quenched with 0.2M HCl(aq), diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile.water, 0.1% TFA) afforded Compound 25. 1HNMR (400 MHz, Chloroform-d) δ 10.44 (s, IH), 8.29 (s, IH), 7.56 - 7.38 (m, 2H), 7.06 (t, J = 9.2 Hz, IH), 5.30 (dd, J = 9.3, 3.5 Hz, IH), 5.21 (s, IH), 4.65 - 4.45 (m, 3H), 4.21 (dd, J = 12.8, 3.4 Hz, IH), 3.95 (dd, J = 12.4, 9.7 Hz, IH), 2.11 - 1.89 (m, 4H), 1.89 1.74 (m, IH), 1.53 (dt, J = 12.4, 3.2 Hz, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C22H19F4N3O5: 482.14; found: 482.2.
Example 26
Préparation of Compound 26 (2S,5R,13aS)-N-(4-chloro-3-fluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Steps 1 and 2
15-B (41 mg, 0.13 mmol) was treated with acetonitrile (I mL), (4-chloro3-fluorophenyl)methanamine (40 mg, 0.25 mmol), HATU (60 mg, 0.16 mmol), andN,Ndiisopropylethylamine (28 mg, 0.22 mmol). The reaction mixture was stirred at room température for one hour and magnésium bromide (48 mg, 0.26 mmol) was added. The mixture was sealed and heated to 50 °C. After 60 minutes, the reaction mixture was quenched with 0.2M HCl (aq), diluted with brine, and thrice extracted into DCM. HPLC purification (Acetonitrile:water, 0.1% TFA) afforded Compound 26. ’H-NMR (400 MHz, Chloroform-d) δ 10.41 (s, IH), 8.30 (s, IH), 7.24 (t, J = 6.1 Hz, IH), 7.13 - 6.90 (m, 2H), 5.30 (dd, J = 9.1, 3.2 Hz, IH), 5.22 (s, IH), 4.61 (s, IH), 4.51 (s, 2H), 4.20 (d, J = 9.4 Hz, IH), 3.95 (d, J = 12.0 Hz, IH), 2.11 - 1.90 (m, 4H), 1.90 - 1.76 (m, IH), 1.53 (d, J = 12.2 Hz, IH). LCMS-EST (m/z)·. [M+H]+calculated for C21H19CIFN3O5: 448.11; found: 448.2.
Example 27
Préparation of Compound 27 (2S,5R)-N-(l-(2,4-difluorophenyl)cyclopropyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l
b] [ 1,3]oxazepine-10-carboxamide
O OH
MgBr2
Step 1
A suspension of the compound 1-A (1.004 g, 3.19 mmol), the amine 275 A (688 mg, 3.35 mmol), and HATU (1.453 g 3.82 mmol) in CH2CI2 (20 mL) was stirred in 0 °C bath as Ν,Ν-diisopropylethylamine (DIPEA) ( 2 mL, 11.48 mmol) was added.
After 1 hour at 0 °C, the reaction mixture was concentrated to a syrup, diluted with ethyl acetate, and washed with water (x 2). After the aqueous fractions were extracted with ethyl acetate (x 1), the organic fractions were combined, dried (ISfeSCU), and 10 concentrated. The residue was purified by CombiFlash (120 g column) using hexanesethyl acetate as eluents. The major peak was combined and concentrated to afford 1.082 g (73%) of the product 27-B. After the minor peak was combined and concentrated, the concentrated residue was dissolved in CH2CI2 and some insoluble materials were filtered. The filtrate was concentrated to get 361 mg (24%) of the additional product 27-B.
LCMS-ESI* (m/z): [M+H]+calculated for C22H25F2N2O7: 467.16; found: 467.1.
Step 2 and 3
Compound 27-B (81 mg, 0.174 mmol) was dissolved in a mixture (1 mL) of acetonitrile (22 mL), AcOH (2 mL), and methanesulfonic acid (0.14 mL, 2.16 mmol) at room température and the resulting solution was stirred at 65 °C for 20 hours.
After the resulting solution was cooled to room température, the aminoalcohol 27-D (50 mg, racemic, 0.363 mmol), K2CO3 (50 mg, 0.362 mmol), and acetonitrile (2 mL) were added to the solution. The resulting mixture was stirred at 65 °C bath for 1 hour. After the reaction mixture was cooled to room température, it was acidified with 1 N HCl (~2 mL), diluted with water (~8 mL), and extracted with CH2CI2 (x 3). Combined extracts were dried (TSfezSCL), concentrated, and purified by CombiFlash to obtain 67 mg (82%) of compound 27-E. ‘H-NMR (400 MHz, CDCh) δ 10.53 (s, IH), 8.25 (s, IH), 7.60 (td, J= 8.5, 6.5 Hz, IH), 6.85 - 6.57 (m, 2H), 5.33 (br, IH), 5.26 (dd, 9.6, 3.9 Hz, IH), 4.60 (t, J= 3.0 Hz, IH), 4.18-4.06 (m, IH), 4.01 (s, 3H), 3.92 (dd, J= 12.7, 9.6 Hz, IH) , 2.11 - 1.91 (m, 4H), 1.88 - 1.71 (m, IH), 1.60 - 1.49 (m, IH), 1.31 - 1.10 (m, 4H). 19F-NMR (376.1 MHz, CDCh) δ -111.80 (q, J= 8.8 Hz, 1F), 112.05 (p, J= 7.9 Hz, 1F). LCMS-ESI+ (m/z): [M+H]+ calculated for C24H24F2N3O5: 472.17; found: 472.1.
Step 4
A mixture of compound 27-E (67 mg, 0.142 mmol) and MgBr2 (66 mg, 0.358 mmol) in MeCN (3 mL) was stirred at 50 °C for 30 minutes and cooled to 0 °C before treating with 1 N HCl (3 mL). After the mixture was diluted with water (—30 mL), the product was extracted with CH2CI2 (x 3), and the combined extracts were dried (Na2SÛ4) and concentrated. The product was purified by préparative HPLC and freezedried to obtain product 27 as a 1:1 mixture with trifluoroacetic acid. ’H-NMR (400 MHz, CDCh) δ 10.70 (s, IH), 8.35 (s, IH), 7.57 (q, J= 8.2 Hz, IH), 6.91 - 6.56 (m, 2H), 5.31 (dt, J= 14.3, 4.0 Hz, 2H), 4.68 (s, IH), 4.22 (dd, J= 13.2, 3.9 Hz, IH), 3.99 (dd, J= 12.8, 9.3 Hz, IH), 2.28 - 1.96 (m, 5H), 1.88 (ddt, J= 12.1, 8.6, 3.7 Hz, IH), 1.71 - 1.49 (m, IH), 1.38 - 1.11 (m, 4H). 19F-NMR (376.1 MHz, CDCh) δ -76.37 (s, 3F), -111.6 ~ -
111.75 (m, 2F). LCMS-ESI+ (m/z): [M+H]+calculated for C23H22F2N3O5:458.15; found: 458.1.
Example 28
Préparation of Compound 28 (2S,6R)-N-( 1 -(2,4-difluorophenyl)cyclopropyl)-9-hydroxy-8,10-dioxo3,4,5,6,8,10,14,14a-octahydro-2H-2,6-methanopyrido[r,2':4,5]pyrazino[2,lb][l,3]oxazocine-l 1-carboxamide
Step 1 and 2
Compound 27-B (87 mg, 0.187 mmol) was dissolved in a mixture (2 mL) of acetonitrile (22 mL), AcOH (2 mL), and methanesulfonic acid (0.14 mL, 2.16 mmol) at room température and the resulting solution was stirred at 65 °C for 20 hours.
After the resulting solution was cooled to room température, the aminoalcohol 28-A (44 mg, racemic, 0.382 mmol) and acetonitrile (2 mL) were added to the solution. After the resulting mixture was stirred at 65 °C bath for 30 minutes, K2CO3 (41 mg, 0.297 mmol) was added and the mixture was stirred at 65 °C for 21 hours. The reaction mixture was cooled to room température, it was acidifîed with 1 N HCl (~2 mL), diluted with water (~8 mL), and extracted with CH2CI2 (x 3). Combined extracts were dried (Na2SO4), concentrated, and purified by préparative HPLC and the fraction containing the product was freeze-dried. After the residue was dissolved in ethyl acetate, the solution was washed with saturated NaHCCL (x 1), dried (Na2SO4), and concentrated to obtain 18 mg (20%) of compound 28-B as a 1:1 mixture with trifluoroacetic acid, ΐ75
NMR (400 MHz, CDCh) δ 10.54 (s, IH), 8.26 (s, IH), 7.63 (td, J= 8.6, 6.6 Hz, IH), 6.76 (dddd, J= 21.9, 11.2, 8.7, 2.3 Hz, 2H), 5.39 (dd, J= 9.6, 3.7 Hz, IH), 4.53-4.36 (m, 2H), 4.09 (dd, J= 12.8, 3.7 Hz, IH), 4.03 (s, 3H), 3.99 (dd, J = 12.7, 9.7 Hz, IH), 2.41-2.20 (m, 2H), 1.84 (dtd, J= 19.7, 9.3, 8.8, 4.4 Hz, 2H), 1.74 (dd, J= 14.6, 2.5 Hz, IH), 1.62 - 1.35 (m,2H), 1.34-1.14 (m, 5H). 19F-NMR (376.1 MHz, CDCh) δ-111.75 (q, J= 8.9 Hz, 1F), -112.01 (p, J- 7.9 Hz, 1F). LCMS-EST (m/z): [M+H]+calculated for C25H26F2N3O5· 486.18; found: 486.2.
Step 3
Compound 28-B (18 mg, 0.037 mmol) was treated with MgBn as described in step4 in the synthesis of compound 27-E to obtain compound 28. 1H-NMR (400 MHz, CDCh) δ 10.66 (s, IH), 8.29 (s, IH), 7.59 (td, J - 8.5, 6.6 Hz, IH), 6.89 6.60 (m, 2H), 5.51 (dd, J= 9.9, 4.0 Hz, IH), 4.55 (s, IH), 4.48 (t, J= 4.2 Hz, IH), 4.21 (dd, J= 12.9, 4.1 Hz, IH), 3.99 (dd, J- 12.8, 9.8 Hz, IH), 2.56 - 2.35 (m, IH), 2.14 (dd, J=16.1, 5.9 Hz, IH), 1.96-1.74 (m, 3H), 1.66 - 1.37 (m, 3H), 1.28 (d, J= 4.4 Hz, 2H), 1.26 - 1.19 (m, 2H). 19F-NMR (376.1 MHz, CDCh) δ -76.41 (s, 3F, -111.79 (m, 2F). LCMS-ESI+ (m/z): [M+H]+ calculated for C24H23F2N3O5: 472.17; found: 472.1.
Example 29
Préparation of Compound 29 (2R,6S)-N-(l-(2,4-difluorophenyl)cyclopropyl)-9-hydroxy-8,10-dioxo3,4,5,6,8,10,14,14a-octahydro-2H-2,6-methanopyrido[l',2':4,5]pyrazino[2,lb][l,3]oxazocine-l 1-carboxamide
Step 1 and 2
Compound 29-B (13 mg, 14%) was prepared from compound 27-B (87 mg, 0.187 mmol) and the aminoalcohol 29-A (45 mg, 0.391 mmol) in a manner similar 5 to that described in step 1 of the synthesis of compound 28-B. ’H-NMR (400 MHz, CDCb) δ 10.54 (s, IH), 8.26 (s, IH), 7.63 (td, J= 8.6, 6.6 Hz, IH), 6.76 (dddd, J- 21.9, 11.2, 8.7, 2.3 Hz, 2H), 5.39 (dd, ./ = 9.6, 3.7 Hz, IH), 4.53 -4.36 (m, 2H), 4.09 (dd, J = 12.8, 3.7 Hz, IH), 4.03 (s, 3H), 3.99 (dd, .7 = 12.7, 9.7 Hz, IH), 2.41 - 2.20 (m, 2H), 1.84 (dtd, .7 = 19.7, 9.3, 8.8, 4.4 Hz, 2H), 1.74 (dd, J- 14.6, 2.5 Hz, IH), 1.62- 1.35 (m, 2H), 10 1.34 - 1.14 (m, 5H). ”F-NMR (376.1 MHz, CDCb) δ -111.75 (q, J = 8.9 Hz, 1F), 112.01 (p, J -7.9 Hz, 1F). LCMS-ESI+ (m/z): [M+H]+calculated for C25H26F2N3O5: 486.18; found: 486.2.
Step 3
Compound 29 was prepared from compound 29-B in a manner similar to 15 that described in step 2 of the synthesis of compound 16. 1H-NMR (400 MHz, CDCb) δ 10.66 (s, IH), 8.29 (s, IH), 7.59 (td, J= 8.5, 6.6 Hz, IH), 6.89 - 6.60 (m, 2H), 5.51 (dd, J= 9.9, 4.0 Hz, IH), 4.55 (s, IH), 4.48 (t, J= 4.2 Hz, IH), 4.21 (dd, J = 12.9, 4.1 Hz, IH), 3.99 (dd,J= 12.8, 9.8 Hz, IH), 2.56-2.35 (m, IH), 2.14(dd, J= 16.1, 5.9 Hz, IH), 1.96- 1.74 (m, 3H), 1.66- 1.37 (m, 3H), 1.28 (d, .7=4,4 Hz, 2H), 1.26-1.19 (m, 2H). 20 19F-NMR (376.1 MHz, CDCb) δ -76.41 (s, 3F, -111.79 (m, 2F). LCMS-ESI+ (m/z): [M+H]+calculated for C24H23F2N3O5: 472.17; found: 472.1.
Example 30
Préparation of Compound 30 (2S,5R,13aS)-N-(l-(2,4-difluorophenyl)cyclopropyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[T,2':4,5]pyrazino[2,l5 b] [ 1,3]oxazepine-10-carboxamide
30-B
O OH
Step 1 and 2
Compound 27-B (150 mg, 0.322 mmol) was dissolved in acetonitrile (2 mL), AcOH (0.2 mL), and methanesulfonic acid (0.007 mL, 0.108 mmol) at room température and the resulting solution was stirred at 65 °C for 20 hours. After the resulting solution was cooled to room température, the aminoalcohol 30-A (72.1 mg, chiral, 0.713 mmol), K2CO3 (89.4 mg, 0.647 mmol), and acetonitrile (2 mL) were added to the solution. The resulting mixture was stirred at 65 °C bath for 0.5 hour. After the reaction mixture was cooled to room température, it was acidified with 1 N HCl (-3 mL), diluted with water (-12 mL), and extracted with CH2CI2 (x 3). Combined extracts were dried (NazSCL), concentrated, and purified by CombiFlash to obtain 128 mg (84%) of compound 30-B. XH-NMR (400 MHz, CDCh) δ 10.52 (s, IH), 8.24 (s, IH), 7.61 (td, J = 8.6, 6.6 Hz, IH), 6.85 - 6.65 (m, 2H), 5.33 (t, J = 4.1 Hz, IH), 5.25 (dd, J = 9.5, 3.9 Hz, IH), 4.61 (d, J = 3.4 Hz, IH), 4.18 - 4.08 (m, IH), 4.02 (s, 3H), 3.99 - 3.87 (m, IH), 2.12 - 1.91 (m, 4H), 1.85 - 1.69 (m, IH), 1.55 (ddd, J = 12.3, 4.1, 2.8 Hz, IH), 1.31 - 1.14 (m, 4H). 19F-NMR (376.1 MHz, CDCh) δ -111.79 (q, J = 8.8 Hz, 1F), -112.05 (p, J = 7.9 Hz, 1F). LCMS-ESr(m/z): [M+H]+calculatedfor C24H24F2N3O5: 472.17; found: 472.2.
Step 3
A mixture of compound 30-B (128 mg, 0.272 mmol) and MgBr2 (130 mg, 0.706 mmol) in MeCN (5 mL) was stirred at 50 °C for 30 minutes and cooled to 0 °C before treating with 1 N HCl (4 mL). After the mixture was diluted with water, the product was extracted with CH2CI2 (x 3), and the combined extracts were dried (NazSO4) and concentrated. The product was purified by CombiFlash to obtain product 30. 1HNMR (400 MHz, CDCh) δ 12.27 (s, IH), 10.52 (s, IH), 8.16 (s, IH), 7.61 (td, J= 8.6,
6.6 Hz, IH), 6.96 - 6.54 (m, 2H), 5.36 - 5.23 (m, 2H), 4.66 (t, 3.1 Hz, IH), 4.18 -
4.06 (m, IH), 3.94 (dd, ./ = 12.8, 9.4 Hz, IH), 2.20- 1.95 (m, 4H), 1.89 (td, J= 11.4, 9.8,
6.7 Hz, IH), 1.70 - 1.54 (m, IH), 1.32 - 1.15 (m, 4H). 19F-NMR (376.1 MHz, CDCh) Ô -111.87 (q, J = 8.9 Hz, 1F), -112.21 (p, J = 7.9 Hz, 1F). LCMS-ESI+ (m/z): [M+H]+ calculated for CssHsîFzNsOs: 458.15; found: 458.2.
Example 31
Préparation of Compound 31 (2R,5S)-N-(l-(2,4-difluorophenyl)cyclopropyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[l',2':4,5]pyrazino[2,1b] [ 1,3 Joxazepine-10-carboxamide
Step 1 and 2
Compound 31-B (123 mg, 81%) was prepared from compound 27-B (150 mg, 0. 322 mmol) and the aminoalcohol 31-A (70.3 mg, 0.695 mmol) in a manner similar to that described in step 1 and 2 of the synthesis of compound 30-B. 1H-NMR (400 MHz, CDCh) δ 10.52 (s, IH), 8.24 (s, IH), 7.62 (td, J= 8.6, 6.6 Hz, IH), 6.91 - 6.63 (m, 2H), 5.33 (t, J = 4.1 Hz, IH), 5.25 (dd, J= 9.5, 3.9 Hz, IH), 4.61 (d, J = 3.4 Hz, IH), 4.14.4.07 (m, IH), 4.03 (s, 3H), 3.93 (dd, <7 = 12.7, 9.5 Hz, IH), 2.12 - 1.91 (m, 4H), 1.85 - 1.69 (m, IH), 1.55 (ddd, .7= 12.3, 4.1, 2.8 Hz, IH), 1.31 - 1.14 (m, 4H). 19F-NMR (376.1MHz, CDCh) δ -111.79 (q, J =9.2, 8.7 Hz, 1F),-112.03 (h, .7=8.1, 7.5 Hz, 1F). LCMS-ESI+ (m/z): [M+H]+ calculated for C24H24F2N3O5: 472.17; found: 472.1.
Step 3
Compound 31 was prepared from compound 31-B in a manner similar to that described in step 3 of the synthesis of compound 30. ’H-NMR (400 MHz, CDCh) δ 12.26 (s, IH), 10.49 (s, IH), 8.13 (s, IH), 7.58 (td, J= 8.6, 6.5 Hz, IH), 6.90 - 6.56 (m, 2H), 5.32 (dd, J= 9.4, 4.1 Hz, IH), 5.27 - 5.22 (m, IH), 4.64 (t, J= 3.1 Hz, IH), 4.11 (dd, J= 12.8, 4.0 Hz, IH), 4.01 - 3.79 (m, IH), 2.28 - 1.95 (m, 4H), 1.95 - 1.80 (m, IH), 1.71 (m, IH), 1.56 (m, IH), 1.42 - 1.08 (m, 4H). 19F-NMR (376.1 MHz, CDCh) δ 80
111.95 (q, J-8 9 Hz, 1F), -112.22 (p, J = 7.9 Hz, IF). LCMS-ESC <Wz): [M+H]* calculated for C23H22F2N3O5: 458.15; found: 458.1.
Example 32
Préparation of Compound 32 (2S,5R)-N-(l-(2,4-difluorophenyl)cyclobutyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5 -methanopyrido [ 1 ',2' :4,5]pyrazino [2,1 -b] [ 1,3 ]oxazepine-10-carboxamide
A solution of compound 32-A (22.2 mg, 0.069 mmol), compound 32-B (18.7 mg, 0.102 mmol), and HATU (43 mg, 0.113 mmol) in CH2CI2 (2 mL) was stirred at room température as Ν,Ν-diisopropylethylamine (DIPEA) (0.075 mL, 0.431 mmol) was added. After 30 minutes, the reaction mixture was diluted with ethyl acetate and washed with water (x 2). After the aqueous fractions were extracted with EA (x 1), the 15 organic fractions were combined, dried, concentrated, and dried in vacuum.
A mixture of the above crude product and MgBr2 (35 mg, 0.190 mmol) in MeCN (2 mL) was stirred at 50 °C bath for 1 hour and cooled to 0 °C before being treated with 1 N HCl (~ 1 mL). The resulting solution was diluted with water, and extracted with
CH2CI2 (x 3). The combined extracts were dried (Na2SO4), and concentrated. The product was purified by préparative HPLC and freeze-dried to obtain compound 32. Î-NMR (400 MHz, CDCh) δ 10.87 (s, IH), ~9.3 (br, IH), 8.35 (s, IH), 7.50 (td, J= 8.7, 6.3 Hz, IH), 6.89 - 6.78 (m, IH), 6.72 (ddd, J= 11.2, 8.9, 2.6 Hz, IH), 5.48 - 5.12 (m, 2H), 4.72 - 4.60 (m, IH), 4.22 (dd, J= 13.0, 4.1 Hz, IH), 3.98 (dd, J= 12.9, 9.4 Hz, IH), 2.68 (m, 4H), 2.33 - 1.98 (m, 6H), 1.90 (m, 2H), 1.60 (ddd, J= 12.4, 4.1, 2.7 Hz, IH). 19F-NMR (376.1MHz, CD3CN) δ -76.39 (s, 3F), -110.50 (q, J= 9.2 Hz, 1F), -112.65 (p, J= 7.8 Hz, 1F). LCMS-ESI+ (m/z): [M+H]+calculated for C24H24F2N3O5: 472.17; found: 472.0.
Example 33
Préparation of Compound 33 (2S,5R)-N-(l-(2,4-difluorophenyl)cyclopentyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb] [ 1,3 ]oxazepine-10-carboxamide
Compound 33 was obtained from compound 32-A and compound 33-A as described in the synthesis of compound 32. ^-NMR (400 MHz, CDCI3) δ 10.70 (s, IH), ~9.5 (br, IH), 8.41 (s, IH), 7.43 (td, J= 8.9, 6.4 Hz, IH), 6.85 - 6.76 (m, IH), 6.72 (ddd, J= 11.5, 8.8, 2.6 Hz, IH), 5.48 - 5.18 (m, 2H), 4.68 (t, J= 3.2 Hz, IH), 4.26 (dd, J = 13.0, 4.1 Hz, IH), 4.00 (dd, J= 13.0, 9.4 Hz, IH), 2.72 - 2.45 (m, 2H), 2.22 - 1.96 (m, 6H), 1.96 - 1.75 (m, 5H), 1.60 (ddd, J = 12.5, 4.1, 2.7 Hz, IH). 19F-NMR (376.1 MHz, CD3CN) δ-76.41 (s, 3F),-107.86 (q, J = 9.4 Hz, 1F),-113.13 (p, J= 8.0 Hz, 1F). LCMS82
ESI+ (m/z): [M+H]+ calculated for C25H26F2N3O5: 486.18; found: 485.9.
Example 34
Préparation of Compound 34 (2S,5R)-N-(l-(2,4-difluorophenyl)cyclohexyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[ 1 *,2' : 4,5]pyrazino[2,1 -b] [ 1,3 ]oxazepine-10-carboxamide
Compound 34 was obtained from compound 32-A and compound 34-A as described in the synthesis of compound 32. ’H-NMR (400 MHz, CDCI3) δ 10.83 (s, IH), ~9.6 (br, IH), 8.44 (s, IH), 7.37 (td, J= 9.0, 6.4 Hz, IH), 6.97 - 6.76 (m, IH), 6.69 (ddd, J= 11.9, 8.8, 2.7 Hz, IH), 5.48 - 5.18 (m, 2H), 4.68 (t, J= 3.0 Hz, IH), 4.28 (dd, J = 13.1, 4.1 Hz, IH), 4.03 (dd, J = 13.0, 9.4 Hz, IH), 2.60 (d, J= 13.1 Hz, 2H), 2.29 - 1.96 (m, 4H), 1.95 - 1.77 (m, 4H), 1.77 - 1.65 (m, 4H), 1.61 (ddd, J= 12.5, 4.1, 2.7 Hz, IH), 1.30 (br, IH). 19F-NMR (376.1 MHz, CD3CN) δ -76.41 (s, 3F), -107.86 (q, J= 9.4 Hz, 1F), -113.13 (p, J = 8.0 Hz, 1F). LCMS-ESI+ (m/z): [M+H]+ calculated for C26H28F2N3O5: 500.20; found: 500.0.
Example 35
Préparation of Compound 35 (2S,5R)-N-(4-(2,4-difluorophenyl)tetrahydro-2H-pyran-4-yl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb] [ 1,3 Joxazepine-10-carboxamide
Compound 35 was obtained from compound 32-A and compound 35-A as 5 described in the synthesis of compound 32. ’H-NMR (400 MHz, CDCh) δ 10.95 (s, IH),
8.33 (s, IH), ~7.6 (br, IH), 7.38 (td, J= 9.0, 6.3 Hz, IH), 6.85 (td, J = 8.4, 2.6 Hz, IH),
6.73 (ddd, J= 11.7, 8.6, 2.6 Hz, IH), 5.32 (dt, J= 14.4, 4.0 Hz, 2H), 4.68 (t, J= 3.1 Hz,
IH), 4.24 (dd, J= 13.0, 3.9 Hz, IH), 4.11 -3.81 (m, 5H), 2.60 (d, J= 13.7 Hz, 2H), 2.33
- 2.17 (m, 2H), 2.18 - 1.97 (m, 4H), 1.87 (m, IH), 1.61 (dt, J= 12.5, 3.3 Hz, IH). 19F10 NMR (376.1 MHz, CD3CN) Ô -76.40 (s, 3F), -108.78 (q, J= 10.3, 9.8 Hz, 1F), -112.63 (p, J= 8.0 Hz, 1F). LCMS-ESI+ (m/z)·. [M+H]+calculated for C25H26F2N3O6: 502.18; found: 502.0.
Example 36
Préparation of Compound 36 (2S,5R)-N-((S)-l-(2,4-difluorophenyl)-2,2,2-trifluoroethyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb] [ 1,3 ]oxazepine-10-carboxamide
Compound 36 was obtained from compound 32-A and compound 36-A as described in the synthesis of compound 32. 1H-NMR (400 MHz, CDCh) δ 11.31 (d, .7= 9.4 Hz, IH), 8.41 (s, IH), 7.65 - 7.44 (m, IH), 6.95 (ddd, J= 9.6, 5.6, 2.0 Hz, IH), 6.92 5 - 6.79 (m, IH), 6.15 (h, J =7.4 Hz, IH), ~6 (br, IH), 5.41 (dd, J= 9.5, 4.0 Hz, IH), 5.31 (t, J = 4.0 Hz, IH), 4.70 (s, IH), 4.34 (dd, J = 12.8, 3.9 Hz, IH), 4.05 (dd, J= 12.9, 9.4 Hz, IH), 2.26 - 1.99 (m, 4H), 1.99 - 1.87 (m, IH), 1.62 (dt, J= 12.6, 3.4 Hz, IH). 19FNMR (376.1 MHz, CDCh) δ -75.23 (t, J = 6.9 Hz, 3F), -76.33 (s, 3F), -108.31 (m, 1F), 112.30 (p, J= 8.0 Hz, 1F). LCMS-ESI+ (m/z): [M+H]+calculated for C22H19F5N3O5: 10 500.12; found: 500.1.
Example 37
Préparation of Compound 37 (3 S, 11 aR)-N-( 1 -(2,4-difluorophenyl)cyclopropyl)-6-hydroxy-3 -methyl-5,7-dioxo2,3,5,7,11,1 la-hexahydrooxazolo[3,2-a]pyrido[l,2-d]pyrazine-8-carboxamide
Step 1
Methyl 5-(l-(2,4-difluorophenyl)cyclopropylcarbamoyl)-l-(2,2dimethoxyethyl)-3-methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (27-B, 0.150 g, 0.32 mmol) in acetonitrile (1.5 mL) and acetic acid (0.2 mL) was treated with methanesulfonic acid (0.05 mL), sealed with a yellow cap, and heated to 70 °C. After 16 hours, the mixture was cooled to afford a crude solution of methyl 5-(1-(2,4difluorophenyl)cyclopropylcarbamoyl)-1 -(2,2-dihydroxyethyl)-3 -methoxy-4-oxo-1,4dihydropyridine-2-carboxylate 27-C. LCMS-ESI* (m/z); [M+H]+ calculated for C18H19F2N2O7: 439; found: 439.
Steps 2 and 3
Methyl 5-(l-(2,4-difluorophenyl)cyclopropylcarbamoyl)-1-(2,2dihydroxyethyl)-3 -methoxy-4-oxo-1,4-dihydropyridine-2-carboxylate (27-C, 0.32 mmol, the crude mixture from the previous step) was dissolved in acetonitrile (1.5 mL) and acetic acid (0.2 mL). (S)-2-aminopropan-l-ol (0.048 g, 0.64 mmol) and K2CO3 (0.088 g, 0.64 mmol) were added to the reaction mixture. The reaction mixture was sealed and heated to 70 °C. After 3 hours, the reaction mixture was cooled and magnésium bromide (0.081 g, 0.44 mmol) was added. The mixture was resealed and heated to 50 °C. After 10 minutes, the reaction mixture was cooled to 0 °C and 1 N hydrochloric acid (0.5 mL) was added in. Then the reaction mixture was diluted with MeOH (2 mL). After filtration, the crude was purified by Prep-HPLC (30-70% acetonitrile:water, 0.1% TFA) to afford Compound 37 as a TFA sait. lH-NMR (400 MHz, Methanol-iA) δ 8.31 (s, IH),
7.62 (td, J= 9.2, 8.7, 6.5 Hz, IH), 7.02 - 6.78 (m, 2H), 5.53 - 5.20 (m, IH), 4.68 (dd, J = 12.3, 4.2 Hz, IH), 4.40 (dq, J= 19.1, 6.7 Hz, 2H), 3.98 (dd, J= 12.2, 10.0 Hz, IH),
3.71 (dd, J= 8.3, 6.3 Hz, IH), 1.41 (d, J = 6.1 Hz, 3H), 1.22 (s, 4H). ”F-NMR (376 MHz, Methanol-ûti) δ -113.66 - -113.95 (m, 1F), -113.94 - -114.29 (m, 1F). LCMS5 ESI+ (m/z): [M+H]+ calculated for C2iH2oF2N305: 432.; found: 432.
Example 38
Préparation of Compound 38 (lS,4R,12aR)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
38-A
H?, Pd/C
NaN-} K H rrr^N3
DMF H Boc 38-D
O OBn
38-G
1) KOH
2) HATU, DIEA
2,4-difluorobenzyiamine
Step 1
A solution of compound 38-A (1562 mg, 5.799 mmol) (see Example 41b in WO 97/05139) in THF (10 mL) was stirred at -78 °C as 2.0 MLiBHk in THF (3.2 mL) was added and the resulting mixture was stirred at room température. After 3 hours, additional 2.0 M L1BH4 in THF (3.2 mL) was added and the solution was stirred at room température for 17.5 hours. After the reaction mixture was diluted with ethyl acetate and added water slowly, two phases were separated, and the separated aqueous fraction was extracted with ethyl acetate (x 1). Two organic fractions were washed with water (x 1), combined, dried (Na2SÛ4), and concentrated. The residue was purified by CombiFlash (40 g column) using hexanes - ethyl acetate as eluents to afford compound 38-B. 1HNMR (400 MHz, Chloroform-d) δ 4.11 (s, IH), 3.65 - 3.52 (m, 2H), 3.45 (m, IH), 2.32 (d, J = 4.1 Hz, IH), 2.20 (s, IH), 1.75 - 1.64 (m, 2H), 1.61 (m, 2H), 1.49 - 1.41 (m, IH), 1.47 (s, 9H), 1.28 - 1.23 (d, J = 10 Hz, IH). LCMS-ESC (m/z): [M+H]+ calculated for C12H22NO3: 228.16; found: 227.7.
Step 2
A solution of compound 38-B (589 mg, 2.591 mmol) and NEt3 (0.47 mL, 3.369 mmol) in CH2CI2 (6 mL) was stirred at 0 °C as MsCl (0.22 mL, 2.842 mmol) was added. After 1 hour at room température, the mixture was diluted with ethyl acetate and washed with water (x 2). The aqueous fractions were extracted with ethyl acetate (x 1), and the organic fractions were combined, dried (Na2SÛ4), and concentrated. The residue was purified by Combi Flash (40 g column) using hexanes - ethyl acetate as eluents to afford compound 38-C. Î-NMR (400 MHz, ChlorofornwZ) δ 4.39 - 4.28 (m, IH), 4.16 (s, 0.4H), 4.06 (s, 0.6H), 3.98 (dd, J= 10.0, 8.7 Hz, 0.6H), 3.86 (t, J= 9.6 Hz, 0.4H), 3.51 (dd, J= 9.3, 3.7 Hz, 0.6H), 3.43 (dd, 9.3, 3.6 Hz, 0.4H), 3.02 (s, 3H), 2.59 (m, IH), 1.82- 1.58 (m, 4H), 1.51 - 1.44 (m, 9H), 1.41 (d,.7= 14.8 Hz, IH), 1.31 (s, 0.6H), 1.29 (s, 0.4H).
Step 3
To a solution of compound 38-C (769 mg, 2.518 mmol,) in DMF (5 mL) was added sodium azide (819 mg, 12.6 mmol). The reaction mixture was stirred at 50 °C for 15 hours, at 80 °C for 5 hours, and at 100 °C for 19 hours. The reaction mixture was diluted with 5% LiCl solution and the product was extracted with ethyl acetate (x 2). After the organic fractions were washed with water (x 1), the two organic fractions were combined, dried (JS^SCU), and concentrated. The residue was purified by CombiFlash (40 g column) using hexanes - ethyl acetate as eluents to afford compound 38-D. *11NMR (400 MHz, Chloroform-zZ) δ 4.16 (s, 0.4H), 4.06 (s, 0.6H), 3.61 (dd, J= 12.2, 3.6 Hz, 0.6H), 3.51 (dd, J= 12.1, 3.2 Hz, 0.4H), 3.38 (dd, .7=9.4, 3.4 Hz, 0.6H), 3.26 (dd, J = 9.8, 3.3 Hz, 0.4H), 3.06 (dd, J= 12.2, 9.4 Hz, 0.6H), 3.01 - 2.92 (m, 0.4H), 2.48 (d, J = 5.2 Hz, IH), 1.82 - 1.57 (m, 4H), 1.46 (d, J= 3.0 Hz, 9H), 1.42 (m, IH), 1.28 (m, 0.6H), 1.27- 1.23 (m, 0.4H).
Step 4
To a solution of compound 38-D (507 mg, 2.009 mmol,) in ethyl acetate (10 mL) and EtOH (10 mL) was added 10% Pd/C (52 mg ). The reaction mixture was stirred under H2 atmosphère for 1.5 hours. The mixture was fîltered through celite and the fîltrate was concentrated to afford crude compound 38-E. LCMS-ESI+ (m/z): [M+H]+calculated for C12H23N2O2: 227.18; found: 226.8.
Step 5
The mixture of crude compound 38-E (206 mg, 0.910 mmol), compound 38-F (330 mg, 0.953 mmol), and NaHCCh (154 mg, 1.833 mmol) in water (3 mL) and EtOH (3 mL) was stirred at room température for 20 hours. After the reaction mixture was diluted with water and extracted with ethyl acetate (x 2), the extracts were washed with water (x 1), combined, dried (Na2SÛ4), and concentrated to afford the crude pyridine product.
The crude residue (388 mg) was dissolved in CH2CI2 (4 mL) and 4 N HCl in dioxane (4 mL). After 1.5 hours, additional 4 N HCl in dioxane (4 mL) was added and stirred for 1 hour at room température. The mixture was concentrated to dryness, coevaporated with toluene (x 1) and dried in vacuum for 30 minutes.
The crude residue and l,8-diazabicycloundec-7-ene (DBU) (1.06 mL, 7.088 mmol) in toluene (10 mL) was stirred at 110 °C bath. After 30 minutes, the mixture was concentrated and the residue was purified by CombiFlash (40 g column) using ethyl acetate - 20% MeOH/ethyl acetate as eluents to obtain compound 38-G. *H-NMR (400 MHz, Chloroform-d) δ 8.03 (s, IH), 7.68 - 7.58 (m, 2H), 7.36 - 7.27 (m, 3H), 5.53 (d, J = 9.9 Hz, IH), 5.11 (d, J = 9.9 Hz, IH), 4.93 (s, IH), 4.43 - 4.30 (m, 2H), 3.89 (dd, J = 12.2, 3.3 Hz, IH), 3.73 (t, J= 12.0 Hz, IH), 3.59 (dd, J = 11.9, 3.3 Hz, IH), 2.53 (d, J =
2.8 Hz, IH), 1.87- 1.67 (m,4H), 1.55 (d,J= 10.0 Hz, IH), 1.51 - 1.45 (m, IH), 1.38 (t, J = 7.1 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+ calculated for C23H25N2O5: 409.18; found: 409.2.
Step 6
The mixture of compound 38-G (232 mg, 0.568 mmol) in THF (3 mL) and MeOH (3 mL) was stirred at room température as 1 N KOH (3 mL) was added. After 1 hour, the reaction mixture was neutralized with 1 N HCl (~3.1 mL), concentrated, and the residue was concentrated with toluene (x 3). After the residue was dried in vacuum for 30 minutes, a suspension ofthe crude residue, 2,4-difluorobenzylamine (86 mg, 0.601 mmol), and HATU (266 mg, 0.700 mmol) were in CH2CI2 (4 mL) and DMF (4 mL) was stirred at 0 °C as Ν,Ν-diisopropylethylamine (DIPEA) (0.7 mL, 4.019 mmol) was added. After 45 minutes, additional 2,4-difluorobenzylamine (86 mg, 0.559 mmol), HATU (266 mg, 0.700 mmol), and Ν,Ν-diisopropylethylamine (DIPEA) (0.7 mL, 4.019 mmol) were added at room température. After 1.25 hours, the mixture was concentrated to remove most of CH2CI2, diluted with ethyl acetate, and washed with 5% LiCl (x 2). After the aqueous fractions were extracted with ethyl acetate (x 1), the organic fractions were combined, dried (Na2SO4), and concentrated. The residue was purified by Combiflash (40 g column) using ethyl acetate -20%MeOH/ethyl acetate as eluents to afford compound 38-H ^-NMR (400 MHz, Chloroform-d) δ 10.48 (t, J = 6.0 Hz, IH), 8.33 (s, IH), 7.62 - 7.51 (m, 2H), 7.40 - 7.27 (m, 4H), 6.87 - 6.75 (m, 2H), 5.39 (d, J = 10.0 Hz, IH), 5.15 (d, J = 10.0 Hz, IH), 4.92 (s, IH), 4.68 - 4.53 (m, 2H), 3.97 (dd, J = 12.5, 3.4 Hz, IH), 3.77 (t, J = 12.2 Hz, IH), 3.55 (dd, J = 12.1, 3.3 Hz, IH), 2.53 (d, J = 3.1 Hz, IH), 1.88 - 1.62 (m, 4H), 1.59 - 1.42 (m, 2H). 19F-NMR (376 MHz, Chloroform-d) δ -112.17 (q, J = 7.6 Hz, 1F), -114.79 (q, J = 8.6 Hz, 1F). LCMS-ESI+ (m/z): [M+H]+ calculated for C28H26F2N3O4: 506.19; found: 506.2.
Step 7
Compound 38-H (240 mg, 0.475 mmol) was dissolved in TFA (3 mL) at room température for 30 minutes, and the solution was concentrated. The residue was purified by CombiFlash (40 g column) using CH2Ch-20% MeOH in CH2CI2 as eluents. After the collected product fractions were concentrated, the residue was triturated in
MeCN (~2 mL) at 0 °C for 15 minutes, and the solids were filtered and washed with MeCN. The collected solids were dried in vacuum to afford compound 38.
The filtrate was concentrated, and the residue was dissolved in MeCN (~1 mL) and water (~1 mL) by heating. The solution was slowly cooled to room température 5 and then in ice bath for 15 minutes. The solids were filtered and washed with MeCN, and dried in vacuum to afford additional compound 38. ’H-NMR (400 MHz, Chloroform-d) δ 11.68 (s, IH), 10.42 (s, IH), 8.27 (s, IH), 7.41 - 7.31 (m, IH), 6.86 - 6.73 (m, 2H), 4.90 (d, J = 2.5 Hz, IH), 4.71 - 4.53 (m, 2H), 4.07 (d, J = 10.6 Hz, IH), 3.90 - 3.67 (m, 2H), 2.68 (s, IH), 2.01 (s, IH), 1.97 - 1.80 (m, 3H), 1.80 - 1.62 (m, 2H). 19F-NMR(376MHz, 10 Chloroform-d) δ -112.28 (m, 1F), -114.74 (m, 1F). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H19F2N3O4: 416.14; found: 416.3.
Examples 39 and 40 Préparation of Compounds 39 and 40 (2R,3S,5R,13aS)-N-(2,4-difluorobenzyl)-8-hydroxy-3-methyl-7,9-dioxo15 2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb][l,3]oxazepine-10-carboxamide 39 and (2S,3R,5S,13aR)-N-(2,4-difluorobenzyl)-8hydroxy-3-methyl-7,9-dioxo-2,3,4,5,7,9,13,13a-octahydro-2,5methanopyrido[ 1 ',2' : 4,5]pyrazino[2,1 -b] [ 1,3 Joxazepine-10-carboxamide 40
MeLi, CuCN
BF3 OEt2
NHBoc
NHBoc
HCI/dioxane,
OH (+/-)
F
Step 1
Cuprous cyanide (290 mg, 3.27 mmol) was suspended in 3.3 mL THF and cooled to -78 °C. A 1.6M solution of MeLi (4.1 mL, 6.56 mmol) in diethyl ether was 5 added dropwise, the reaction solution was allowed to warm to room température over the course of 2 hours, and recooled to -78 °C. Tert-butyl (lR,3R,5S)-6oxabicyclo[3.1.0]hexan-3-ylcarbamate (330 mg, 1.66 mmol) was added dropwise in 3.3 mL THF, followed by boron trifluoride diethyl etherate (0.25 mL, 1.99 mmol), allowed to warm to -30 °C over 30 minutes, and stirred between -35 °C and -25 °C for one hour.
The reaction solution was then warmed to room température and quenched with a mixture of saturated NH3(aq)/NH4(aq), extracted to EtOAc, washed with brine, dried over MgSO4, filtered, concentrated, and purified by SGC (0-10% EtOH/DCM) to afford racemic tert-butyl (lS,3S,4S)-3-hydroxy-4-methylcyclopentylcarbamate. *H-NMR (400 MHz, Chloroform-ri) δ 5.16 (s, IH), 3.98 (s, IH), 3.74 (q, J= 4.3 Hz, IH), 3.65 (q, J = 15 7.0 Hz, IH), 2.23 (dt, J= 14.0, 7.0 Hz, IH), 1.98 (dt, J= 13.3, 7.0 Hz, IH), 1.89 - 1.79 (m, IH), 1.58 - 1.44 (m, IH), 1.38 (s, 9H), 1.18 (t, J= 7.0 Hz, IH), 0.91 (d, 7.0 Hz,
3H).
Step 2 mL HCl/dioxane (4M, 12 mmol) was added to a solution of racemic tertbutyl (lS,3S,4S)-3-hydroxy-4-methylcyclopentylcarbamate (182 mg, 0.85 mmol) in 3 mL dioxane. The reaction mixture was stirred at room température for 2 hours, concentrated and twice chased with toluene to afford racemic (lS,2S,4S)-4-amino-2methylcyclopentanol.
Step 3
Methyl 5-(2,4-difluorobenzylcarbamoyl)-l-(2,2-dihydroxyethyl)-3methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 310 mg, 0.75 mmol), racemic (lS,2S,4S)-4-amino-2-methylcyclopentanol (115 mg, 0.76 mmol), and potassium carbonate (232 mg, 1.68 mmol) were taken up in 3.8 mL acetonitrile/0.2 mL acetic acid and stirred at 90 °C for 2 hours, after which the reaction mixture was partitioned between DCM and brine, the aqueous phase extracted to DCM, combined organic phases dried over MgSÛ4, filtered, concentrated, and purified by SGC (Or 10% EtOH/DCM) to afford intermediate 39-A.
Step 4
Intermediate 39-A (190 mg) was separated by chiral Prep-HPLC on a Lux Cellulose-2 column using 9:1 ACNLMeOH as eluent to afford Intermediates 39-B (first eluting peak) and 40-A (second eluting peak) in enantioenriched form. For intermediate 39-B: (absolute stereochemistry confirmed by XRay crystallography), Chiral HPLC rétention time = 3.98 minutes (Lux Cellulose-2 IC, 150 x 4.6 mm, 2 mL/min 9:1 ACN:MeOH). For intermediate 40-A: (absolute stereochemistry confirmed by XRay crystallography), Chiral HPLC rétention time = 6.35 minutes (Lux Cellulose-2 IC, 150 x 4.6 mm, 2 mL/min 9:1 ACN:MeOH).
Step 5 a
Magnésium bromide (68 mg, 0.37 mmol) was added to a solution of intermediate 39-B (83 mg, 0.18 mmol) in 2 mL acetonitrile. The reaction mixture was stirred at 50 °C for 1 hour, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted to dichloromethane. The combined organic phases were dried over MgSO4, filtered, concentrated, and purified by silica gel chromatography (0-10% EtOH/DCM) to afford compound 39. 1H-NMR (400 MHz, Chloroform-d) δ 12.32 (s, IH), 10.36 (s, IH), 8.29 (s, IH), 7.44 - 7.33 (m, IH), 6.88 - 6.76 (m, 2H), 5.37 (dd, J = 9.5, 4.1 Hz, IH), 5.28 (t, J = 5.3 Hz, IH), 4.63 (d, J =
5.9 Hz, 2H), 4.23 (d, J = 23.0 Hz, 2H), 3.99 (dd, J = 12.7, 9.5 Hz, IH), 3.72 (q, J = 7.0 Hz, IH), 2.51 (dq, J= 13.7, 6.8, 6.1 Hz, IH), 2.15 (ddd, J= 14.7, 8.3, 2.3 Hz, IH), 1.94 (d, J = 12.7 Hz, IH), 1.77 (ddd, J = 12.7, 4.0,2.9 Hz, IH), 1.61 (dt, J = 14.6, 5.2 Hz, 2H), 1.24 (t, J = 7.0 Hz, IH), 1.09 (d, J = 7.2 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+ calculated for C22H22F2N3O5: 446.15; found: 446.2.
Step 5b
Magnésium bromide (59 mg, 0.32 mmol) was added to a solution of intermediate 40-A (70 mg, 0.15 mmol) in 2 mL acetonitrile. The reaction mixture was stirred at 50 °C for 1 hour, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted to dichloromethane. The combined organic phases were dried over MgSO4, filtered, concentrated, and purified by silica gel chromatography (0-10% EtOH/DCM) to afford compound 40. 'H-NMR (400 MHz, Chloroform-ri) δ 12.32 (s, IH), 10.36 (s, IH), 8.29 (s, IH), 7.44 - 7.33 (m, IH), 6.88 - 6.76 (m, 2H), 5.37 (dd, J= 9.5, 4.1 Hz, IH), 5.28 (t, J= 5.3 Hz, IH), 4.63 (d, J =
5.9 Hz, 2H), 4.23 (d, 23.0 Hz, 2H), 3.99 (dd, J= 12.7, 9.5 Hz, IH), 3.72 (q, J = 7.0
Hz, IH), 2.51 (dq, J= 13.7, 6.8, 6.1 Hz, IH), 2.15 (ddd, ./ = 14.7, 8.3, 2.3 Hz, IH), 1.94 (d,J= 12.7 Hz, IH), 1.77 (ddd, J= 12.7, 4.0, 2.9 Hz, IH), 1.61 (dt,J= 14.6, 5.2 Hz, 2H), 1.24 (t, J= 7.0 Hz, IH), 1.09 (d, J= 7.2 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+calculated for C22H22F2N3O5: 446.15; found: 446.2.
Example 41
Préparation of Compound 41 (lR,4S,12aR)-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[l ,2-a: 1 ',2'-d]pyrazine-9-carboxamide
Boc
41-A
NatL DMF
MsCI ,NaHCO3
2) HCl
3) DBU
Step 1
A solution of the 41-A (2020 mg, 7.463 mmol) (prepared by the same method as 38-A) in THF (14 mL) was stirred at 0 °C as 2.0 M LiBH4 in THF (7.5 mL, 15 mmol) was added. After the resulting mixture was stirred at rt for 21 h, it was cooled at 10 0 °C and diluted with EA before water was added slowly to quench. After two phases were separated, the aqueous fraction was extracted with EA (x 1) and the two organic fractions were washed with water (x 1), combined, dried (NaîSCL), and concentrated. The residue was purified by CombiFlash (120 g column) using hexanes EA as eluents to get 41-B. LCMS-ESF (œ/z): [M-C4Hg+H]+calculated for CsHuNCh: 172.10; found: 171.95.
Step 2
A 100-mL round bottom flask was charged with reactant 41-B (1.6 g, 7.05 mmol) and triethylamine (0.94 g, 9.3 mmol) in DCM (20 mL). Methanesulfonyl chloride (0.91 g, 8.0 mmol) was added to the reaction mixture. Then the reaction mixture was stirred at room température for 3 hours. The mixture was diluted with EA (100 mL) and washed with water (2x). The aqueous fractions were extracted with EA (Ix), and the organic fractions were combined, dried (Na2SO4), and concentrated. The residue was purified by Combi Flash (120 g column, cartridge used) using hexanes - EA as eluents to afford 41-C LCMS-ESL (m/z): [M+H]+calculated for CîsHisFsNzCfr 306; found: 306.
Step 3
A 100-mL round bottom flask was charged with reactant 41-C (2.1 g, 6.9 mmol) and sodium azide (2.3 g, 34.5 mmol) in DMF (10 mL). Then the reaction mixture was stirred at 100 °C for ovemight. The mixture was diluted with EA (100 mL) and washed with water (2x). The aqueous fractions were extracted with EA (lx), and the organic fractions were combined, dried (TfeSCh), and concentrated. The residue was purified by Combi Flash (120 g column, cartridge used) using hexanes - EA as eluents to afford 41-D LCMS-ESI+ (m/z): [M+H]+calculated for C18H19F2N2O7: 253; found: 253.
Step 4
To a solution (purged with N2) of reactant 41-D (1.3 g) in EA (20 mL) and EtOH (20 mL) was added Pd/C (130 mg). The mixture was stirred under H2 for 3 hours. The mixture was filtered through celite and the filtrate was concentrated to afford compound 41-E LCMS-ESI+ (m/z): [M+H]+ calculated for C18H19F2N2O7: 227; found: 227.
Step 5
A 100-mL round bottom flask was charged with reactant 41-E (1.05 g, 4.62 mmol) and reactant 38-F (1.6 g, 4.62 mmol) in Ethanol (20 mL). Sodium bicarbonate (0.77 g, 9.2 mmol) in water (20 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température ovemight. The mixture was diluted with EA (100 mL) and washed with water (2x). The aqueous fractions were extracted with EA (lx), and the organic fractions were combined, dried (Na2SO4), and concentrated. The crude product (2.4 g) was used for next step without further purification. LCMS-ESI+ (m/z): [M+H]+calculated for C18H19F2N2O7: 556; found: 556.
A 100-mL round bottom flask was charged with the crude product from the previous reaction in 4 N HCl /dioxane (24.7 mL). Then the reaction mixture was stirred at room température for 1 hour. After concentration, the intermediate (2.1 g) and DBU (3.27 g, 21.5 mmol) in toluene (30 mL) was heated to 110 °C with stirring for 1 hour. After concentration, the residue was purified by CombiFlash (120 g column) using hexanes - ethyl acetate as eluents to afford 41-F. LCMS-ESI+ (m/z): [M+H]+ calculated for C18H19F2N2O7: 409; found: 409.
Step 6
A 100-mL round bottom flask was charged with reactant 41-F (0.5 g, 1.22 mmol) in THF (5 mL) and MeOH (5 mL). 1 N KOH (3.7 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for 1 hour. The reaction mixture was acidified by adding 1 N HCl (3.7 mL), concentrated to remove most of organic solvents, and extracted with EtOAc (2 X). The organic layers were combined, dried (Na2SC>4), and concentrated to afford compound 41-G.
Step 7
A 100-mL round bottom flask was charged with reactant 41-G (0.14 g, 0.37 mmol), (2,4,6-trifluorophenyl)methanamine (0.12 g, 0.73 mmol), Ν,Νdiisopropylethylamine (DIPEA) (0.24 g, 1.84 mmol) and HATU (0.28 g, 0.74 mmol) were dissolved in DCM (5 mL). The reaction mixture was stirred at room température for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHCOs (2x), saturated NH4C1 (2x) and dried over Na2SO4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford compound 41-H. LCMS-EST+ (m/z): [M+H]+ calculated for C18H19F2N2O7: 524.5; found: 524.5.
Step 8
A 50-mL round bottom flask was charged with reactant 41-H (0.13 g, 0.25 mmol) in TFA (2 mL). The reaction mixture was stirred at room température for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 41. ’H-NMR (400 MHz, Chloroform-d) δ
11.61 (s, IH), 10.70 - 10.01 (m, IH), 8.26 (s, IH), 6.65 (t, J = 8.1 Hz, 2H), 4.88 (s, IH), 10 4.65 (dd, J = 6.1, 2.4 Hz, 2H), 4.07 (d, J = 10.9 Hz, IH), 3.93 - 3.58 (m, 2H), 2.67 (d, J =
3.1 Hz, IH), 2.08 - 1.41 (m, 7H). ”F-NMR (376 MHz, Chloroform-d) δ -109.22 (d, J =
11.6 Hz, 1F), -111.04 - -112.79 (m, 2F). LCMS-ESI+ (m/z): [M+H]+calculated for C21H20F2N3O5: 434.; found: 434.
Example 42
Préparation of Compound 42 (2R, 5 S, 13 aR)-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorobenzyi)-2,3,4,5,7,9,13,13 aoctahydro-2,5 -methanopyridof 1 ',2' :4, SJpyrazino [2,1 -b] [ 1,3 Joxazepine-10-carboxamide
Step 1
-(2,2-dimethoxyethy 1)-5-methoxy-6-(methoxycarbonyl)-4-oxo-1,45 dihydropyridine-3-carboxylic acid (3.15 g, 10 mmol) in acetonitrile (36 mL) and acetic acid (4 mL) was treated with methanesulfonic acid (0.195 mL, 3 mmol) and placed in a 75 deg C bath. The reaction mixture was stirred for 7 h, cooled and stored at-10 °C for 3 days and reheated to 75 °C for an additional 2 h. This material was cooled and carried on crude to the next step.
Step 2
Crude reaction mixture from step 1 (20 mL, 4.9 mmol) was transferred to a flask containing (lR,3S)-3-aminocyclopentanoI (0.809 g, 8 mmol). The mixture was diluted with acetonitrile (16.8 mL), treated with potassium carbonate (0.553 g, 4 mmol) 15 and heated to 85 °C. After 2 h, the reaction mixture was cooled to ambient température and stirred ovemight. 0.2M HCl (50 mL) was added, and the clear yellow solution was extracted with dichloromethane (2x150 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated to 1.49 g of a light orange solid.
Recrystallization from dichlormethane:hexanes afforded the desired intermediate 42A:
LCMS-ESI+ (m/z): [M+H]+ calculated for C15H17N2O6: 321.11; found: 321.3.
Step 3
Intermediate 42-A (0.225 g, 0.702 mmol) and (2,4,6trifluorophenyl)methanamine (0.125 g, 0.773 mmol) were suspended in acetonitrile (4 mL) and treated with Ν,Ν-diisopropylethylamine (DIPEA) (0.183 mmol, 1.05 mmol). To this suspension was added (dimethylamino)-2V,A-dimethyl(3Ær-[l,2,3]triazolo[4,5ô]pyridin-3-yloxy)methaniminium hexafluorophosphate (HATU, 0.294 g, 0.774 mmol). After 1.5 hours, the crude reaction mixture was taken on to the next step. LCMS-ESU (m/z): [M+H]+calculated for C22H21F3N3O5: 464.14; found: 464.2.
Step 4
To the crude reaction mixture of the previous step was added MgBr2 (0.258 g, 1.40 mmol). The reaction mixture was stirred at 50 °C for 10 minutes, acidifîed with 10% aqueous HCl, and extract twice with dichloromethane. The combined organic phases were dried over MgSCM, filtered, concentrated, and purified by silica gel chromatography (EtOH/dichlormethane) followed by HPLC (ACN/H2O with 0.1% TFA modifier) to afford compound 42: 1H-NMR(400 MHz, DMSO-dé) δ 12.43 (s, IH), 10.34 (t, J= 5.7 Hz, IH), 8.42 (s, IH), 7.19 (t, J= 8.7 Hz, 2H), 5.43 (dd, J= 9.5, 4.1 Hz, IH), 5.08 (s, IH), 4.66 (dd, J- 12.9, 4.0 Hz, IH), 4.59 (s, IH), 4.56-4.45 (m, 2H), 4.01 (dd, J= 12.7, 9.7 Hz, IH), 1.93 (s, 4H), 1.83 (d, J= 12.0 Hz, IH), 1.56 (dt, J= 12.0, 3.4 Hz, IH). LCMS-ESI4 (m/z): [M+H]+ calculated for C21H19F3N3O5: 450.13; found: 450.2.
Example 43
Préparation of Compound 43 (12aR)-N-((R)-1 -(2,4-difluorophenyl)ethyl)-7-hydroxy-6,8-dioxo-1,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: r,2'-d]pyrazine-9-carboxamide
100
43-A
Step 1
A 100-mL round bottom flask was charged with reactant 41-G (0.14 g,
0.37 mmol), (R)-l-(2,4-difluorophenyl)ethanamine (0.12 g, 0.74 mmol), N,Ndiisopropylethylamine (0.24 g, 1.84 mmol) and HATU (0.28 g, 0.74 mmol) and were dissolved in DCM (5 mL). The reaction mixture was stirred at room température for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHCCL 10 (2x), saturated NH4CI (2x) and dried over Na2SÛ4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford compound 43-A. LCMS-ESI+ (m/z): [M+H]+calculated for CigH^lWy: 520; found: 520.
Steps 2
A 50-mL round bottom flask was charged with reactant 43-A (0.14 g, 0.27 15 mmol) in TFA (2 mL). The reaction mixture was stirred at room température for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 43. ’H-NMR (400 MHz, Chloroform-d) δ 11.65 (s, IH), 10.57 (s, IH), 8.22 (s, IH), 7.31 (m, IH), 6.99 - 6.62 (m, 2H), 5.64 - 5.32 (m, IH), 4.90 (d, J = 2.7 Hz, IH), 4.04 (d, J = 11.5 Hz, IH), 3.93 - 3.63 (m, 2H), 2.67 (s,
101
IH), 2.08 -1.40 (m, 9H). 19F-NMR (376 MHz, Chloroform-d) δ-113.09 (m, 1F),-115.01 (m, 1F). LCMS-ESI+ (m/z): [M+H] * calculated for C21H20F2N3O5: 430.; found: 430.
Example 44
Préparation of Compound 44 (13aS)-8-hydroxy-7,9-dioxo-N-(2,3,4-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro2,5-methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Step 1
Compound 15-B (40 mg, 0.12 mmol) was taken up in 1 mL acetonitrile and treated with 2,3,4-trifluorobenzylamine (29 mg, 0.18 mmol), HATU (53 mg, 0.14 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (20 mg, 0.16 mmol), and stirred at room température for 2 hours, after which LCMS analysis revealed complété consumption of compound 15-B and formation of intermediate 44-A. The reaction mixture was carried onto the next step.
Step 2
To the crude reaction solution of the previous step was added MgBn (63 mg, 0.34 mmol). The reaction mixture was stirred at 50 °C for one hour, acidified with
102
10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted to dichloromethane. The combined organic phases were dried over MgSCh, filtered, concentrated, and purified by HPLC (ACN/H2O with 0.1% TFA modifier) to compound 44. *H-NMR (400 MHz, DMSO-dô) δ 12.45 (s, IH), 10.38 (t, J = 6.0 Hz, IH), 8.43 (s, IH), 7.27 (q, J = 9.2 Hz, IH), 7.16 (q, J = 8.5 Hz, IH), 5.42 (dd, J= 9.5, 4.0 Hz, IH), 5.08 (s, IH), 4.76 - 4.47 (m, 4H), 4.01 (dd, J= 12.8, 9.7 Hz, IH), 1.92 (s, 4H), 1.82 (d, J= 12.1 Hz, IH), 1.55 (dt, J = 12.2, 2.9 Hz, IH). LCMS-ESI+ (m/z): [M+H]+calculated for C21H19F3N3O5: 450.13; found: 450.2.
Example 45
Préparation of Compound 45 (13aS)-8-hydroxy-7,9-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,4,5,7,9,13,13a-octahydro2,5-methanopyrido[T,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Step 1
Compound 15-B (38 mg, 0.12 mmol) was taken up in 1 mL acetonitrile and treated with 2,4,6-trifluorobenzylamine (34 mg, 0.21 mmol), HATU (50 mg, 0.13 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (23 mg, 0.18 mmol), and stirred at room température for 2 hours, after which LCMS analysis revealed complété consumption of
103 compound 15-B and formation of intermediate 45-A. The reaction mixture was carried onto the next step.
Step 2
To the crude reaction solution of the previous step was added MgBn (55 mg, 0.30 mmol). The reaction mixture was stirred at 50 °C for one hour, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgSO4, filtered, concentrated, and purified by HPLC (ACN/H2O with 0.1% TFA modifier) to afford compound 45. ’H-NMR (400 MHz, DMSO-Ts) δ 12.37 (s, IH), 10.37 - 10.25 (m, IH), 8.37 (s, IH), 7.14 (t, J = 8.7 Hz, 2H), 5.37 (dd, J = 9.5, 4.0 Hz, IH), 5.02 (s, IH), 4.66-4.40 (m, 4H), 3.95 (dd,J=12.8, 9.6 Hz, IH), 1.87 (s, 4H), 1.77 (d, J = 11.9 Hz, IH), 1.50 (dt, J= 11.8, 3.2 Hz, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H19F3N3O5: 450.13; found: 450.2.
Example 46
Préparation of Compound 46 (13aS)-N-(2,6-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13a-octahydro-2,5methanopyrido[r,2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
104
Step 1
Compound 15-B (38 mg, 0.12 mmol) was taken up in 1 mL acetonitrile and treated with 2,6-difluorobenzylamine (19 mg, 0.14 mmol), HATU (56 mg, 0.15 5 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (20 mg, 0.15 mmol), and stirred at room température for 90 minutes, after which LCMS analysis revealed complété consumption of compound A and formation of intermediate 46-A. The reaction mixture was carried onto the next step.
Step 2
To the crude reaction solution of the previous step was added MgBr2 (50 mg, 0.27 mmol). The reaction mixture was stirred at 50 °C for one hour, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgSÛ4, filtered, concentrated, and purified by HPLC (ACN/H2O with 0.1% TFA modifier) to afford compound 46. ’H-NMR (400 MHz, DMSO-îZs) δ 12.37 (s, IH), 10.33 - 10.26 (m, IH), 8.37 (s, IH), 7.39 - 7.29 (m, IH), 7.05 (t, J= 7.9 Hz, 2H), 5.37 (dd, J = 9.5, 4.1 Hz, IH), 5.02 (s, IH), 4.66 - 4.45 (m, 4H), 3.95 (dd, J= 12.7, 9.6 Hz, IH), 1.87 (s, 4H), 1.77 (d, J= 12.0 Hz, 1H), 1.50 (dt, J= 12.2, 3.5 Hz, IH). LCMS-ESr (m/z): [M+H]+calculated for C2iH2oF2N305: 432.14; found: 432.2.
105
Example 47
Préparation of Compound 47 (lR,4S,12aR)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido [ 1,2-a: 1 ',2'-d]pyrazine-9-carboxamide
41-G 47-A
Step 1
The crude acid 41-G (0.45 g, 1.18 mmol), 2,4-difluobenzylamine (0.35 g,
2.44 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.79 g, 6.11 mmol) and HATU (0.93 g, 2.44 mmol) were dissolved in DCM (10 mL). The reaction mixture was stirred at room température for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHCCh (2x), saturated NLLCl (2x) and dried over Na2SO4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford compound 47-A. LCMS-ESI+ (m/z): [M+H]+ calculated for C18H19F2N2O7.· 506; found: 506.
106
Step 2
A 50-mL round bottom flask was charged with reactant 47-A (0.5 g, 0.99 mmol) in TFA (6 mL). The reaction mixture was stirred at room température for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 47. 1H NMR (400 MHz, Chloroform-d) δ 11.70 (s, IH), 10.44 (s, IH), 8.29 (s, IH), 7.60 - 7.29 (m, IH), 6.95 - 6.58 (m, 2H), 4.10 (s, IH), 4.02 - 3.54 (m, 3H), 2.68 (d, J = 3.1 Hz, IH), 2.00 - 1.40 (m, 8H). 19F NMR (376 MHz, Chloroform-d) δ -112.31 (d, J = 8.0 Hz, 1F), -114.77 (d, J = 8.4 Hz, 1F). LCMSESI+ (m/z): [M+H]+ calculated for C21H20F2N3O5: 416.; found: 416.
Example 48
Préparation of Compound 48 (lS,4R,12aS)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[l ,2-a: 1 ',2'-d]pyrazine-9-carboxamide
48-B was prepared analogously to 55-H in Example 55, substituting 48A for 55-A. Compound 48 was prepared as described for compound 38 in Example 38, substituting 48-B for 38-B to afford compound 48. 1H-NMR (400 MHz, Chloroform-d) δ 11.79 (s, IH), 10.44 (m, IH), 8.33 (s, IH), 7.42 - 7.31 (m, IH), 6.86 - 6.74 (m, 2H), 4.74 (s, IH), 4.63 (d, J = 5.8 Hz, 2H), 4.19 (m, IH), 4.07 - 4.03 (m, 2H), 2.83 (s, IH),
107
1.92 - 1.68 (m, 6H). 19F NMR (376 MHz, Chloroform-d) δ -112.3 (m, 1F), -114.8 (m, 1F). LCMS-ESI+ (m/z): [M+H]+calculated for C21H20F2N3O4: 416.14.; found: 416.07.
Example 49
Préparation of Compound 49 (2S,5R,13aS)-8-hydroxy-7,9-dioxo-N-((3-(trifluoromethyl)pyridin-2-yl)methyl)2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb] [ 1,3 ] oxazepine-10-carboxamide
Step 1
Compound 15-B (44 mg, 0.14 mmol) was taken up in 1 mL acetonitrile and treated with (3-(trifluoromethyl)pyridin-2-yl)methanamine (38 mg, 0.18 mmol, HCl 15 sait), HATU (69 mg, 0.18 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.07 mL, 0.40 mmol), and stirred at room température for 1 hour, after which LCMS analysis revealed complété consumption of compound 15-B and formation of intermediate 49-A. The reaction mixture was carried onto the next step.
108
Step 2
To the crude reaction solution of the previous step was added MgBr2 (51 mg, 0.28 mmol). The reaction mixture was stirred at 50 °C for 90 minutes, acidified with
10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgSCfi, filtered, concentrated, and triturated by methanol followed by diethyl ether to afford compound 49. 1H-NMR (400 MHz, DMSO-ûfc) δ 12.42 (s, IH), 10.80 - 10.70 (m, IH), 8.83 (d, J-5.0 Hz, lH),8.44(s, IH), 8.19 (d, J= 8.6Hz, IH), 7.56 (dd, 7.7,
5.2 Hz, IH), 5.43 (dd, J= 9.5, 4.0 Hz, IH), 5.08 (s, IH), 4.86 - 4.80 (m, 2H), 4.67 (dd, J 10 - 12.9, 4.0 Hz, IH), 4.59(s, IH), 4.02 (dd, J= 12.6, 9.8 Hz, IH), 1.93 (s, 4H), 1.82 (d, J = 12.1 Hz, IH), 1.60 - 1.52 (m, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C2iH2oF3N405: 465.14; found: 465.2.
Examples 50 and 51
Préparation of Compounds 50 and 51
N-(2,4-difluorobenzyl)-9-hydroxy-8,10-dioxo-2,3,5,6,8,10,14,14a-octahydro-2,6methanopyrido[r,2':4,5]pyrazino[2,l-b][l,6,3]dioxazocine-ll-carboxamide 50 and 51
109
OH
Chiral SFC
IC column
51
Step 1
Methyl 5-(2,4-difluorobenzylcarbamoyl)-l-(2,2-dihydroxyethyl)-3methoxy-4-oxo-l,4-dihydropyridine-2-carboxylate (1-C, 392 mg, 0.95 mmol) (Example
87), racemic cA-5-aminotetrahydro-2H-pyran-3-ol (WO 2012/145569 Bennett, B. L. et al, filed April 20, 2012 ) (112 mg, 0.95 mmol), and potassium carbonate (134 mg, 0.97 mmol) were taken up in 3.8 mL acetonitrile/0.2 mL acetic acid and stirred at 90 °C for 90 minutes, after which the reaction mixture was partitioned between DCM and brine, the aqueous phase extracted with DCM, combined organic phases dried over MgSÛ4, 10 filtered, concentrated, and purified by SGC (0-10% EtOH/DCM) to afford intermediate
50-A.
Step 2
Intermediate 50-A (40 mg) was separated by chiral SFC on a Chiralpak
IC column using 10% DMF in supercritical carbon dioxide as eluent to afford 15 Intermediates 50-B (first eluting peak) and 51-A (second eluting peak) in enantioenriched form. For intermediate 50-B: (absolute stereochemistry unknown), Chiral HPLC rétention time = 11.48 minutes (Chiralpak IC, 150 x 4.6 mm, 1 mL/min MeOH). For
110 intermediate 51-A: (absolute stereochemistry unknown), Chiral HPLC rétention time = 14.35 minutes (Chiralpak IC, 150 x 4.6 mm, 1 mL/min MeOH).
Step 3 a
Magnésium bromide (12 mg, 0.06 mmol) was added to a solution of intermediate 50-B (10.5 mg, 0.02 mmol, absolute stereochemistry unknown) in 1 mL acetonitrile. The reaction mixture was stirred at 50 °C for 1 hour, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgSCU, filtered, concentrated, and purified by HPLC (ACN/H2O with 0.1% TFA modifier) to afford compound 50. ’H-NMR (400 MHz, Chloroform-7) δ 10.47 (t, J= 5.8 Hz, IH), 8.42 (s, IH), 7.35 (q, J = 8.6, 8.2 Hz, IH), 6.81 (q, J= 8.7, 8.0 Hz, 2H), 6.41 (dd, 7 = 10.0, 3.6 Hz, IH), 4.79 (s, IH), 4.65 (s, 2H), 4.36 - 4.26 (m, 2H), 4.20 - 4.08 (m, 2H), 3.98 (dd, 7= 12.4, 10.2 Hz, IH), 3.88 (t, 7= 11.8 Hz, 2H), 2.27 (dt, 7= 13.3, 3.1 Hz, IH), 2.15 - 2.06 (m, IH). LCMS-ESI+ (m/z); [M+H]+ calculated for C21H20F2N3O6: 448.40; found: 448.2.
Step 3b
Magnésium bromide (13 mg, 0.07 mmol) was added to a solution of intermediate 51-A (13.2 mg, 0.03 mmol, absolute stereochemistry unknown) in 1 mL acetonitrile. The reaction mixture was stirred at 50 °C for 1 hour, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgSCL, filtered, concentrated, and purified by HPLC (ACN/H2O with 0.1% TFA modifier) to afford compound 51. *H-NMR (400 MHz, Chloroform-d) δ 10.47 (t, J— 5.8 Hz, IH), 8.42 (s, IH), 7.35 (q, 7 = 8.6, 8.2 Hz, IH), 6.81 (q, J= 8.7, 8.0 Hz, 2H), 6.41 (dd, 7= 10.0, 3.6 Hz, IH), 4.79 (s, IH), 4.65 (s, 2H), 4.36 - 4.26 (m, 2H), 4.20 - 4.08 (m, 2H), 3.98 (dd, 7= 12.4, 10.2 Hz, IH), 3.88 (t, 7= 11.8 Hz, 2H), 2.27 (dt, 7= 13.3, 3.1 Hz, IH), 2.15 - 2.06 (m, IH). LCMS-ESff (m/z); [M+H]+ calculated for C21H20F2N3O6: 448.40; found: 448.2.
111
Exampie 52
Préparation of Compound 52 (2S,5R,13aS)-N-(2-cyclopropoxy-4-fluorobenzyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l
b] [ 1,3 Joxazepine-10-carboxamide
Step 1
A solution cyclopropanol (1.9 g, 29 mmol) in 20 mL dioxane was added dropwise to a 0 °C solution of Sodium hydride (60% dispersion in minerai oil, 1.04 g, 26 mmol) in 80 mL dioxane. The reaction mixture was allowed to warm to room température, 2,4-difluorobenzonitrile (3.48 g, 25 mmol) was added portionwise, and reaction température raised to 95 °C. The reaction solution was cooled to room 15 température after stirring for 18 hours, diluted with ethyl acetate, washed twice with water and twice with brine, dried over MgSOy filtered, and concentrated onto silica gel. Purification by silica gel chromatography (0-10% EtOAc/hexanes) afforded 2cyclopropoxy-4-fluorobenzonitrile. 1H-NMR (400 MHz, Chloroform-d) δ 7.52 (dd, J =
112
8.6, 6.2 Hz, IH), 7.05 (dd, J= 10.5, 2.3 Hz, IH), 6.73 (td, J = 8.2, 2.3 Hz, IH), 3.87 3.76 (m, IH), 0.87 (m, 4H).
Step 2
To a 0 °C suspension of lithium aluminum hydride in THF (IM, 15 mL, 15 mmol) was added 2-cyclopropoxy-4-fluorobenzonitrile in 14 mL diethyl ether dropwise. The reaction solution was stirred for 3 hours, gradually warming to room température, at which point it was recooled to 0 °C, an additional 8 mL lithium aluminum hydride in THF (IM, 8 mmol) added, and stirred for an additional 90 minutes. The reaction was quenched by sequential addition of 0.9 mL water, 0.9 mL 15% NaOH(aq), and 2.7 mL water. The reaction was filtered through celite with diethyl ether rinses, dried over MgSCk, and concentrated to afford 2-cyclopropoxy-4~fluorobenzylamine of sufficient purity to carry on as crude. ’H-NMR (400 MHz, Chloroform-d) δ 7.17 - 7.08 (m, IH), 6.96 (dd, J= 10.9, 2.4 Hz, IH), 6.61 (td, J= 8.3, 2.5 Hz, IH), 3.78 - 3.66 (m, 3H), 0.89-0.72 (m, 4H).
Step 3
Compound 15-B (46 mg, 0.14 mmol) was taken up in 1 mL acetonitrile and treated with 2-cyclopropoxy-4-fluorobenzylamine (32 mg, 0.18 mmol), HATU (62 mg, 0.16 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.04 mL, 0.22 mmol), and stirred at room température for 2 hours, after which LCMS analysis revealed complété consumption of compound 15-B and formation of intermediate 52-A. The reaction mixture was carried onto the next step.
Step 4
To the crude reaction solution of the previous step was added MgBn (56 mg, 0.30 mmol). The reaction mixture was stirred at 50 °C for 90 minutes, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgSCri, filtered, concentrated, and purified by HPLC (ACNÆhO with 0.1% TFA modifier) to afford compound 52.1H-NMR(400 MHz, DMSO-iZs) δ 12.44 (s, IH), 10.21 (t, J= 5.8 Hz, IH), 8.41 (s, IH), 7.22 - 7.15 (m, IH), 7.12 (dd, J = 11.2, 2.5 Hz, IH),
113
6.72 (td, J= 8.5, 2.5 Hz, IH), 5.42 (dd, J= 9.6, 4.1 Hz, IH), 5.07 (s, IH), 4.66 (dd, J = 12.8, 4.1 Hz, IH), 4.58 (s, IH), 4.34 (dd, J= 5.6, 2.4 Hz, 2H), 4.04 - 3.91 (m, 2H), 1.92 (s, 4H), 1.82 (d, J= 11.9 Hz, IH), 1.55 (dt, J= 12.4, 3.5 Hz, IH), 0.80 (q, J= 6.3, 5.7 Hz, 2H), 0.72 (q, J = 6.0, 4.9 Hz, 2H). LCMS-ESI+ (m/z): [M+H]+ calculated for C24H25FN3O6: 470.17; found: 470.1.
Example 53
Préparation of Compound 53 (2R,5S,13aR)-N-(2-cyclopropoxy-4-fluorobenzyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb] [ 1,3 Joxazepine-10-carboxamide
Step 1
Compound 42-A (46 mg, 0.14 mmol) was taken up in 1 mL acetonitrile and treated with 2-cyclopropoxy-4-fluorobenzylamine (33 mg, 0.18 mmol), HATU (61 mg, 0.16 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.04 mL, 0.24 mmol), and stirred at room température for 2 hours, after which LCMS analysis revealed complété consumption of compound 42-A and formation of intermediate 53-A. The reaction mixture was carried onto the next step.
114
Step 2
To the crude reaction solution of the previous step was added MgBr2 (55 mg, 0.30 mmol). The reaction mixture was stirred at 50 °C for 90 minutes, acidified with 10% aqueous HCl, partitioned between the aqueous and dichloromethane, and the aqueous phase extracted with dichloromethane. The combined organic phases were dried over MgSCfo filtered, concentrated, and purified by HPLC (ACN/H2O with 0.1% TFA modifier) to afford compound 53.1H-NMR (400 MHz, DMSO-üfc) δ 12.44 (s, IH), 10.21 (t, J = 5.8 Hz, IH), 8.41 (s, IH), 7.22 - 7.15 (m, IH), 7.12 (dd, J = 11.2, 2.5 Hz, IH), 6.72 (td, J= 8.5, 2.5 Hz, IH), 5.42 (dd, J= 9.6, 4.1 Hz, IH), 5.07 (s, IH), 4.66 (dd, J = 12.8, 4.1 Hz, IH), 4.58 (s, IH), 4.34 (dd, J= 5.6, 2.4 Hz, 2H), 4.04 - 3.91 (m, 2H), 1.92 (s, 4H), 1.82 (d, J= 11.9 Hz, IH), 1.55 (dt, J= 12.4, 3.5 Hz, IH), 0.80 (q, J = 6.3, 5.7 Hz, 2H), 0.72 (q, J = 6.0, 4.9 Hz, 2H). LCMS-ESI+ (m/z): [M+H]+ calculated for C24H25FN3O6: 470.17; found: 470.1.
Example 54
Préparation of Compound 54 (2R,5S)-N-((S)-l-(2,4-difluorophenyl)-2,2,2-trifluoroethyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,l
b] [ 1,3 Joxazepine-10-carboxamide
115
54-A
Step 1
A 50-mL round bottom flask was charged with reactant 54-A (0.02 g, 0.06 mmol), (S)-l-(2,4-difluorophenyl)-2,2,2-trifluoroethanamine (0.019 g, 0.09 mmol), N,Ndiisopropylethylamine (DIPEA) (0.048 g, 0.38 mmol) and HATU (0.036 g, 0.09 mmol) in DCM (2 ml). The reaction mixture was stirred at room température for 1 hour. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 mL), washed with saturated NaHCO3 (2x), saturated NH4CI and dried over Na2SÛ4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 54-B LCMS-ESH (m/z): [M+H]+calculated for C18H19F2N2O7: 514; found: 514.
Step 2
A 50-mL round bottom flask was charged with reactant 54-B (0.03 g, 0.058 mmol) and magnésium bromide (0.03 g, 0.15mmol) in acetonitrile (2 mL). The reaction mixture was heated to 50 °C. After 10 minutes, the reaction mixture was cooled to 0 °C and 1 N hydrochloric acid (0.5 mL) was added in. Then the reaction mixture was diluted with MeOH (2 mL). After filtration, the crude was purified by Pre-HPLC purification (30-70% acetonitrile:water, 0.1% TFA) afforded compound 54 as TFA sait. Î-NMR (400 MHz, Chloroform-d) δ 11.28 (d, J = 9.4 Hz, IH), 8.39 (s, IH), 7.54 (q, J = 7.8 Hz, IH), 7.12 - 6.76 (m, 2H), 6.40 - 5.98 (m, IH), 5.57 - 5.18 (m, 2H), 4.68 (s, IH), 4.29 (dd, J= 13.1, 4.0 Hz, IH), 4.05 (dd, J= 12.9, 9.3 Hz, IH), 2.39 - 1.94 (m, 4H), 1.86 (t, J = 10.5 Hz, IH), 1.60 (dt, J = 12.6, 3.4 Hz, IH). 19F-NMR(376 MHz, Chloroform-d) δ -75.30 (t, J = 6.8 Hz, 3 F), -108.33 (dd, J = 8.6, 6.3 Hz, 1F), -111.56 - -113.23 (m, 1 F). LCMS-ESI+ (m/z): [M+H]+calculated for C21H20F2N3O5: 500.; found: 500.
116
Example 55
Préparation of Compound 55 (lR,4S,12aS)-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)-l,2,3,4,6,8,12,12aoctahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
1) H2, Pd(OH)2/C
2) HCI/EtOH r.t.
Boc2O
----.---------------------gs»
NaOH, dioxane r.t., ovemight
bh3-dms
55-G
THF
Step 1
117
A mixture of compound 55-A (40.60 g, 150 mmol) and Pd(OH)2/C (12 g) in EtOH (400 mL) under an atmosphère of H2 was stirred at room température ovemight. The reaction mixture was filtered and treated with HCl/EtOH (400 ml). The mixture was stirred at room température for 2 h. The reaction mixture was concentrated to give compound 55-B, which was used in next step without purification. LCMS-ESI+ (m/z): [M+H]+calculated for C9H16NO: 170.1.; found: 170.2.
Step 2
To a solution of compound 55-B (92.25 g, 0.45 mol) and K2CO3 (186.30 g, 1.35 mol) in CH3CN (1 L) was added benzyl bromide (76.50 g, 0.45 mol) at 0 °C. The mixture was stirred at room température ovemight. The reaction mixture was filtered, concentrated and the residue was purified by chromatography on silica gel to give compound 55-C.
Step 3
To a mixture of diisopropylamine (50 g, 0.50 mol) in THF (400 mL) was added n-BuLi (200 mL, 0.50 mol) at -78 °C at N2 atmosphère. After 0.5 h, the reaction mixture was warmed to 20 °C and stirred for 0.5 h. The mixture was cooled to -78 °C and added a solution of compound 55-C (64.75 g, 0.25 mol) in THF (600 mL) under N2 atmosphère. The mixture was stirred for 4 h and quenched with saturated NH4CI solution. The mixture was extracted with EtOAc and the organic layer was washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by chromatography on silica gel to give compound 55-D
Step 4
A mixture of compound 55-D (129.50 g 0.50 mol) in 4N HCl (1.30 L) was refluxed for 4 h. the mixture was concentrated. The residue was purified by HPLC to give compound 55-E.
118
Step 5
To a mixture of compound 55-E (47 g, 176 mmol) and Pd(OH)2/C (9 g) in EtOH (400 mL) under an atmosphère of H? was stirred at room température ovemight.
The reaction mixture was concentrated to give compound 55-F, which was used in next step without purification. *H-NMR (400 MHz, CDCh) δ 4.22 (s, IH), 4.06 (s, IH), 2.982.95 (d,./= 11.2 Hz, IH), 1.96-1.93 (d, J= 11.2 Hz, IH), 1.86-1.82 (m, 2H), 1.76-1.74 (d, J= 9.2 Hz, 2H), 1.49 (s, IH). LCMS-ESE (m/z): [M+Hf calculated for C7H12NO2:
142.1. ; found: 142.1.
Step 6
To a mixture of compound 55-F (29.20 g, 165 mmol) and 2N NaOH solution (330 mL, 0.66 mol) in dioxane (120 mL) was added Boc2O (39.60 g, 181 mmol) at 0 °C. The reaction mixture was stirred at room température ovemight. The mixture was 15 adjusted with 3 N HCl to pH=5~6 and extracted with DCM. The organic layer was dried over Na2SC>4, filtered and concentrated to give 55-G. 1H-NMR (400 MHz, CDCI3) δ 4.40 (s, IH), 4.26 (s, lH),2.89(s, IH), 1.76-1.74 (s, IH), 1.69-1.59 (m, 4H), 1.50 (s, IH), 1.47 (s, 9H). LCMS-ESI+ (m/z): [M+Na]+calculated for CnHigNNaCU: 264.1.; found:
264.1.
Step 7
To a mixture of compound 55-G (500 mg, 2.07 mmol) in THF (10 mL) chilled to 0 °C was added BH3-DMS THF complex (2N in THF, 8.23 mmol, 4.1 mL) slowly. Gas évolution occured. Internai température was monitored to ensure no major 25 exotherm. Reaction was allowed to warm to r.t. ovemight. Some starting material remained by LC/MS, additional 2 mL BH3-DMS THF complex was added and the mixture was stirred for additional 3 hr then cooled reaction to 0 °C and slowly quenched with methanol (gas évolution occurs). Internai température monitored to ensure exotherm below 25 °C. The mixture was concentrated then purified by silica gel 30 chromotography (20-40% EtOAc/Hexanes) to afford 55-H.
119
Step 8
Compound 55 was prepared as described for Example 41, substituting 55H for 41-B to afford compound 55. ’H-NMR (400 MHz, DMSO-d6) δ 11.81 (s, IH), 10.40 (t, J= 5.8 Hz, IH), 8.39 (s, IH), 7.19 (t, J = 8.6 Hz, 2H), 4.59-4.48 (m, 4H), 4.16 (t, J - 12.2 Hz, IH), 4.03 (d, J = 12.2 Hz, IH), 2.69 (s, IH), 1.75 (d, J = 10.1 Hz, IH),
1.69 - 1.55 (m, 5H). 19F NMR (376 MHz, DMSO-d6) δ -109.3 (m, 1F), -112.5 (m, 1F). LCMS-ESr (m/z): [M+H]+calculated for C21H19F3N3O4: 434.13.; found: 434.32.
Example 56
Préparation of Compound 56 (lR,2S,4R,12aR)-2-fluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)l,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:l',2'-d]pyrazine-9carboxamide
120
H
56-B 56-C
1) H2 Pd/C
2) BOC2O,
56-A
56-G
Step 1
A solution of 56-A (5 g, 19.43 mmol) in tetrahydrofuran (65 ml) was cooled in an ice bath as 0.5 M 9-borabicyclo[3.3.1]nonane (48.58 ml) was added dropwise. The reaction mixture was warmed up to room température. After 18 hours, the reaction was cooled to 0 °C and a mixture of 2M sodium hydroxide (34 ml) and hydrogen peroxide (9.34 ml, 97.15 mmol) was added dropwise. After 2 hours at 0 °C, the reaction was warmed up to room température and stirred for 1 hour. The mixture was diluted with EtOAc and washed with water. The aqueous fractions were extracted with EtOAc, and 10 the organic fractions combined were dried (Na2SO4) and concentrated. The residue was purified by silica column chromatography (50-70% EtOAc/hexanes) to afford 56-B (3.05 g, 57%). LCMS-ESI+ (m/z): [M+H]+ calculated for C16H21NO3: 275.34; found: 276.122.
121
Step 2
To a solution of 56-B (1.45 g, 5.27 mmol) in N,N-dimethylformamide (12 ml) was added tert-butylchlorodiphenylsilane (1.51 ml, 5.79 mmol) and imidazole (1.08 g, 15.8 mmol). After 18 hours, the mixture was diluted with water, extracted into EtOAc (2x), the organic fractions were combined, dried (Na2SO4), and concentrated. The residue was purified by silica column chromatography (10-20% EtOAc/hexanes) to afford 56-C (2.6 g, 96.1%). LCMS-ESr (m/z): [M+H]+ calculated for C32H39NO3Si: 513.74; found: 514.625.
Step 3
To a solution of 56-C (3.27 g, 6.36 mmol) in EtOH (26 mL) and acetic acid (3 mL) was added 10% PdOH/C (0.52 g, 3.7 mmol) and the suspension was shaken in a Parr apparatus at 50 atm for 20 hours. After filtering through Celite, the cake was washed with EtOH, the filtrate was concentrated under vacuum. The residue was dissolved in éthanol (26 ml) and acetic acid (3 ml, 52.4 mmol), treated with 10% PdOH/C (0.52 g, 3.7 mmol) and shaken in a Parr apparatus at 50 atm for 20 hours. Filtered through Celite, the cake was washed with EtOH, the filtrate was concentrated under vacuum to dryness to afford the crude deprotected product (2.07g, 79.4 %). LCMS-ESI+ (m/z): [M+H]+ calculated for C24H31NO3S1: 409.59; found: 410.485.
To the crude residue (2 g, 4.88 mmol) and di-tert-butyl dicarbonate 97% (2.14 g, 9.79 mmol) in THF (20 ml) was added Ν,Ν-diisopropylethylamine (DIPEA) (2.14 ml, 12.27 mmol). After 20 h, the reaction mixture was diluted with water, extracted into EtOAC (2x) and the two organic fractions were washed with water, combined, dried (Na2SO4), and concentrated. The residue was purified by silica column chromatography (10-20% EtOAc/Hexanes) to afford 56-D (2.13 g, 86.14%). LCMS-ESF (m/z): [M+H]T calculated for C30H41NO5S1: 523.74; found: 523.922.
Step 4
A solution of 56-0 (2.07 g, 4.06 mmol) in THF (20 ml) was stirred in an ice bath as 2.0 M L1BH4 in THF (4.07 ml) was added and the resulting mixture was stirred at room température for 18 h. After, the reaction mixture was diluted with ethyl acetate and treated slowly with water. The two phases were separated, and the aqueous fraction
122 was extracted again with ethyl acetate. The two organic fractions were washed with water, combined, dried (NaaSCU), and concentrated. The residue was purified by silica column chromatography (20-40% EOAc/hexanes) to afford 56-E (1.59 g, 81.3%). LCMS-ESI+ (m/z): [M+H]+ calculated for C28H39NO4S1: 481.7; found: 482.337.
Step 5
A mixture of 56-E (1.58 g, 3.28 mmol), phthalimide (0.79 g, 5.38 mmol) and triphenylphosphine (1.93 g, 7.37 mmol) in THF (90 ml) was cooled in an ice bath. Diisopropyl azodicarboxylate, 95% (1.46 ml, 7.42 mmol) was added. The mixture was then warmed up to room température and stirred for 20 h. After, the reaction mixture was concentrated and the residue dissolved in ether, cooled in an ice bath and stirred for 1.5 h. The solids were filtered off and the filtrate was concentrated. The residue was purified by silica column chromatography (10-30% EtOAc/hexanes) to afford the protected amino compound (1.86 g, 92.8%).
A solution of the protected amino compound 56-F (1.85 g, 3.03 mmol) and hydrazine hydrate (0.6 ml, 12.39 mmol) in éthanol (19 ml) was stirred at 70 °C for 2 h The reaction mixture was cooled in an ice bath, ether (10 ml) was added and the mixture was stirred for 30 min. The solid formed was filtered off and the filtrate was concentrated under vacuum to dryness.
Step 6
A mixture of crude amino compound 56-F (991 mg, 2.06 mmol), compound 38-F (Example 38) (714 mg, 2.06 mmol) and NaHCCh (347 mg, 4.12 mmol) in water (15 mL) and EtOH (15 mL) was stirred for 20 h. The reaction mixture was concentrated under vacuum and the residue was partitioned between water and EtOAc. The aqueous layer was re-extracted with EtOAc and the combined organic layers were dried (Na2SO4) and concentrated. The residue (1.5 g) was dissolved in CH2CI2 (5 mL) and 4N HCl in dioxane (18.6 mL) was added. After 1.5 hours the mixture was concentrated to dryness, co-evaporated with toluene and dried in vacuo.
The crude residue (1.38 g) and DBU (1.4 ml, 9.38 mmol) in toluene (25 ml) was stirred at 110 °C. After 35 minutes the mixture was concentrated and the residue was purified by silica column chromatography (5-15% MeOH/EtOAc) to afford 56-G
123 (450 mg, 72.3%). LCMS-ESI+ (m/z): [M+H]+ calculated for C39H42N2O6S1: 662.85; found: 663.766.
Step 7
The mixture of 56-G (890 mg, 1.34 mmol) in MeOH (14 ml) and THF (14 ml) was stirred at room température as IM KOH (7.09 ml) was added. After 30 min the reaction mixture was neutralized with IN HCl, extracted into EtOAc (2x) and the combined organic extracts were dried (Na2SO4) and concentrated.
A suspension of the crude residue (850 mg), 2,4,6-trifluorobenzylamine (248 mg, 1.54 mmol) and HATU (662 mg, 1.74 mmol) in dichloromethane (5 ml) was stirred at room température asΝ,Ν-diisopropylethylamine (DIPEA) (1.63 ml, 9.37 mmol) was added. After 1 h, additional 2,4,6-difluorobenzylamine (32 mg, 0.2 mmol), HATU (153 mg, 0.4 mmol) and Ν,Ν-diisopropylethylamine (DIPEA) (0.12 ml, 0.67 mmol) were added. After 30 minutes the mixture was diluted with water, extracted into EtOAc (3x) the combined organic phases were dried (Na2SO4), concentrated and the residue was purified by silica column chromatography (50-75% EtOAc/hexanes) to afford 56-H (919 mg, 88.23%). LCMS-EST (m/z): [M+H]+ calculated for C44H42F3N3O5S1: 777.9; found: 778.409.
Step 8
A solution of 56-H (915 mg, 1.18 mmol) in THF (5 ml) was stirred in an ice bath as 1.0 M tetrabutylammonium fluoride in THF (1.18 ml) was added dropwise. The resulting mixture was stirred at room température for 30 min. The reaction mixture was concentrated under vacuum and the residue was diluted with EtOAc, washed with water, dried (Na2SO4), concentrated and the residue was purified by silica column chromatography (50-75% EtOAc/hexanes then 5% MeOH/EtOAc). The resulting material (248 mg, 0.46 mmol) was dissolved in dichloromethane (2 ml) cooled to -78°C as diethylaminosulfur trifluoride (0.07 mL, 0.55 mmol) was added dropwise and the reaction was warmed to room température and stirred for 1 h. The reaction was cooled in an ice bath and quenched with saturated NaHCO3, two phases were separated, and the separated aqueous fraction was extracted with CH2CI2. The two organic fractions were combined dried (Na2SÛ4), and concentrated. The residue was purified by silica column
124 chromatography (1% MeOH/EtOAc) to afford 56-J (75 mg) (LCMS-ESI+ (m/z): [M+H]+ calculated for C28H23F4N3O4: 541.49; found: 542.320) and 56-1 (30 mg) (LCMS-ESF (m/z): [M+H]+ calculated for C28H22F3N3O4: 521.49; found: 522.05).
Step 9
Compound 56-J (75 mg, 139 mmol) was dissolved in TFA (1 mL), stirred at room température for 10 minutes, and the solution was concentrated. The residue was purified by reverse phase HPLC (Gemini, 15 to 43% ACN/H2O + 0.1% TFA) to afford compound 56. ’H-NMR (400 MHz, DMSO-d6) δ 10.67 (s, IH), 7.80 (s, IH), 7.17 (t, J = 8.6 Hz, 2H), 5.45-5.18 (m, IH), 4.70-4.39 (m, 3H), 4.23 (d, J= 11.5 Hz, IH), 4.11 - 3.85 (m, 2H), 2.85 (dd, J = 4.2, 2.0 Hz, IH), 2.34 - 2.13 (m, IH), 1.81 (s, IH), 1.55 1.33 (m, 2H). 19F-NMR (376 MHz, DMSO-76) δ -74.20 (m), -106.95 - -116.45 (m), 190.65--194.54 (m).
Example 57
Préparation of Compound 57 (lR,4R,12aR)-2,2-difluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)l,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9carboxamide
125
Step 1
A solution of 57-A (1.45 g, 5.34 mmol) in dichloromethane (30 ml) was cooled in an ice bath as Dess Martin periodinane (4.53 g, 10.69 mmol) was added in portions and the reaction was stirred at room température for 18 h. The reaction was quenched by addition of water, the precipitate was fîltered off and a saturated solution of Na2S2Û3 was added. The mixture was stirred until the biphasic solution turned then saturated NaHCOs was added and the aqueous layer extracted with CH2CI2. The combined organic fractions were dried (Na2SO4) and concentrated. The residue was 10 purified by silica column chromatography (30-50% EtOAc/Hexanes) to afford 57-B (1.13 g, 78.2 %). LCMS-ESI+ (m/z): [M+H]+ calculated for C13H19NO5: 269.29; found: 269.722.
Step 2
A solution of 57-B (0.5 g, 1.86 mmol) in dichloromethane (10 ml) was cooled to -78°C as diethylaminosulfur trifluoride (0.52 mL, 3.91 mmol) was added dropwise and the reaction was warmed to room température and stirred for 18 h. The
126 reaction was cooled in an ice bath and quenched with saturated NaHCCh, two phases were separated, and the separated aqueous fraction was extracted with CH2CI2. The two organic fractions were combined, dried (Na2SÛ4) and concentrated. The residue was purified by silica column chromatography (20-50% EtOAc/hexanes) to afford 57-C (518 mg, 95.39%). 1H-NMR (400 MHz, Chloroform-d) δ 4.43 (s, IH), 4.36 - 4.27 (m, IH), 4.22 (s, IH), 3.75 (s, 3H), 2.95 (t, J = 8.1 Hz, IH), 2.30 - 1.98 (m, 2H), 1.85 - 1.71 (m, IH), 1.44 (m, 9H).
Step 3
A solution of 57-C (935 mg, 3.21 mmol) in THF (10 ml) was stirred in an ice bath as 2.0 M LiBH4 in THF (3.22 ml) was added and the resulting mixture was stirred at room température for 18 h. After, the reaction mixture was diluted with ethyl acetate and water was added slowly. The two phases were separated, and the separated aqueous fraction was extracted with ethyl acetate. The two organic fractions were washed with water, combined, dried (NaîSO.-i), and concentrated. The residue was purified by silica column chromatography (20-40% EtOAc/hexanes) to afford 57-D (724 mg, 85.67%). Î-NMR (400 MHz, Chloroform-J) δ 4.30 - 3.48 (m, 5H), 2.75 - 2.56 (m, IH), 2.24 - 1.90 (m, 3H), 1.86 - 1.65 (m, IH), 1.47 (s, 9H).
Step 4
A mixture of 57-D (720 mg, 2.74 mmol), phthalimide (402 mg, 2.73 mmol) and triphenylphosphine (1.61 g, 6.15 mmol) in THF (45 ml) was cooled in an ice bath. Diisopropyl azodicarboxylate, 95% (1.22 ml, 6.19 mmol), was added. The mixture was then warmed up to room température and stirred for 20 h. After, the reaction mixture was concentrated and the residue dissolved in ether, cooled in an ice bath and stirred for 1.5 h. After the solids were filtered off, the fîltrate was concentrated. The residue was purified by silica column chromatography (40-60% EtOAc/hexanes) to afford the phthalimide adduct (1.07 g, 99.7%). LCMS-ESC (m/z): [M+H]+ calculated for C20H22F2N2O4: 392.4; found: 393.204
A solution of the phthalimide adduct (1.07 g, 2.73 mmol) and hydrazine hydrate (0.54 mL, 11.15 mmol) in éthanol (10 ml) was stirred at 70 °C for 2 hours The reaction mixture was cooled in an ice bath and ether (10 ml) was added. The mixture was
127 stirred for 30 min. The solid formed was filtered off and the filtrate was concentrated under vacuum to dryness to afford crude 57-E.
Step 5
A mixture of crude 57-E (709 mg, 2.7 mmol) compound 38-F (Example 38) (936 mg, 2.7 mmol) and NaHCCE (454 mg, 5.41 mmol) in water (15 mL) and EtOH (15 mL) was stirred for 20 h. The reaction mixture was concentrated under vacuum and the residue was partitioned between water and EtOAc. The aqueous layer was reextracted with EtOAc and the combined organic layers were dried (Na2SO4) and concentrated. The residue (1.5 g) was dissolved in CH2CI2 (7 mL) and 4N HCl in dioxane (26.9 mL) was added. After 1.5 hours the mixture was concentrated to dryness, coevaporated with toluene and dried in vacuum. The crude residue (1.3 g) and DBU (2 ml, 13.4 mmol) in toluene (25 ml) was stirred at 110 °C. After 35 minutes the mixture was concentrated and the residue was purified by silica column chromatography (5-15% MeOH/EtOAc) to afford 57-F (426 mg, 36.17%). LCMS-ESI+ (m/z); [M+H]+ calculated for C23H22F2N2O5: 444.43; found: 445.280.
Step 6
The mixture of compound 57-F (426 mg, 0.96 mmol) in MeOH (7 ml) and THF (7 ml) was stirred at room température as IM KOH (5.06 ml) was added. After 30 minutes the reaction mixture was neutralized with IN HCl, extracted into EtOAc (2x) and the combined organic extracts were dried (Na2SÛ4) and concentrated to crude 57-G. Step 7
A suspension of the crude residue 57-G (189 mg), 2,4,6trifluorobenzylamine (95 mg, 0.59 mmol) and HATU (276 mg, 0.73 mmol) in dichloromethane (3 ml) was stirred at room température as N,N-diisopropylethylamine (DIPEA) (0.59 ml, 3.4 mmol) was added. After 1 h he mixture was diluted with water, extracted into EtOAc (3x). The combined organic phases were dried (Na2SÛ4) and concentrated to 57-H. LCMS-ESU (m/z); [M+H]+ calculated for C28H22F5N3O4: 559.48; found: 560.24.
128
Step 8
Compound 57-H (150 mg, 0.27 mmol) was dissolved in TFA (2 mL), stirred at room température for 10 min, and the solution was concentrated. The residue was purified by reverse phase HPLC (Gemini, 15 to 60% ACN/H2O + 0.1% TFA), to afford compound 57 (85 mg, 67.5%). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H16F5N3O4: 469.36; found: 470.229. Î-NMR (400 MHz, DMSO-î/6) δ 10.41 (t, J = 5.6 Hz, IH), 8.20 (s, IH), 7.12 (t, J= 8.7 Hz, 2H), 4.79 (s, IH), 4.48 (m, 3H), 4.10 (m, 2H), 3.02 (d, J= 5.7 Hz, IH), 2.33 (m, IH), 2.22 - 1.97 (m, 2H), 1.85 (d, J= 11.0 Hz, IH), 1.21 (s, IH). 19F NMR (376 MHz, DMSCMs) δ -69.88 , -71.77 , -74.09 , -88.33 (dd, J= 222.6, 23.8 Hz), -109.15 - -109.60 (m), -110.04 , -112.44 (t, J= 7.6 Hz).
Example 58
Préparation of Compound 58 (lR,4R,12aR)-N-(3-chloro-2,4-difluorobenzyl)-2,2-difluoro-7-hydroxy-6,8-dioxo1,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a: l',2'-d]pyr azine-9carboxamide
129
Step 1
A suspension of the crude residue 57-G (120 mg), 3-chloro,2,4difhiorobenzylamine (67 mg, 0.38 mmol) and HATU (175 mg, 0.46 mmol) in dichloromethane (3 ml) was stirred at room température as N,N-diisopropylethyIamine (DIPEA) (0.38 ml, 0.28 mmol) was added. After 1 h the mixture was diluted with water, extracted into EtOAc (3x) the combined organic phases were dried (Na2SÛ4) and concentrated to yield 58-A. LCMS-EST (m/z): [M+H]+ calculated for C28H22CIF4N3O4: 575.94; found: 576.394.
Step 2
Compound 58-A (166 mg) was dissolved in TFA (2 mL), stirred at room température for 10 min, and the solution was concentrated. The residue was purified by reverse phase HPLC (Gemini, 15 to 70% ACN/H2O + 0.1% TFA), to afford compound 57 (60 mg, 42.8%). LCMS-EST (m/z): [M+H]+ calculated for C21H16CIF4N3O4: 485.82; found: 486.135. *H-NMR (400 MHz, DMSO-d6) δ 10.77 (t, J = 6.0 Hz, IH), 7.77 (s, IH), 7.28 (m, 2H), 4.77 (s, IH), 4.64 - 4.40 (m, 2H), 4.27 (d, J = 9.1 Hz, IH), 3.93 (m, 2H), 2.95 (d, J = 5.8Hz, IH), 2.51 (s, IH), 2.42-2.17 (m, IH), 2.14-1.89 (m, 2H), 1.77 (m, IH). 19F-NMR(376 MHz, DMSO-ùfc) δ -87.63 , -88.23 , -108.67 , -109.27 , -116.42 (t, J = 7.0 Hz), -118.48 (d, 7.8 Hz).
Example 59
Préparation of Compound 59 (lR,2R,4R,12aR)-2-fluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)l,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:T,2'-d]pyrazine-9carboxamide
130
1) KOH
2) HATU, DEA,
2,4,6-F3BnNH2
RSiO
F
1) TBAF
2) Dast
OEt
59-F
59-G
Step 1
A solution of 57-B (1.9 g, 7.06 mmol) in methanol (35 mL) was stirred at 5 0 °C as sodium borohydride (667 mg, 17.64 mmol) was added portionwise and the resulting mixture was stirred at room température for 30 min The reaction mixture was cooled in an ice bath quenched by addition of water and concentrated. The residue was partitioned between water and EtOAc. The aqueous layer was re-extracted with EtOAc and the combined organic layers were dried (Na2SO4) and concentrated The residue was 10 purified by silica column chromatography (30-60% EtOAc/hexanes) to afford 59-A (1.49
g). ‘H-NMR (400 MHz, chloroform-t/) δ 4.57 (s, IH), 4.52 - 4.42 (m, 2H), 4.28 (s, IH), 4.14 (s, IH), 3.72 (d, J = 2.1 Hz, 3H), 2.74 (s, IH), 2.08 - 1.87 (m, 2H), 1.43 (d, J = 23.1 Hz, 10H) and 57-A (96 mg): 1H-NMR (400 MHz, chloroform-J) δ 4.65 - 4.40 (m, 2H), 4.34 - 4.02 (m, IH), 3.73 (d, J = 2.3 Hz, 3H), 2.74 (t, J = 5.3 Hz, IH), 2.12 - 1.55 (m, 15 3H), 1.52 -1.18 (m, 11H).
Step 2
To a solution of 59-A (686 mg, 2.53 mmol) in N,N-dimethylformamide
131 (5 ml) was added tert-butylchlorodiphenylsilane (0.723 mL, 2.78 mmol) and imidazole (516 mg, 7.56 mmol). After 18 h, the mixture was diluted with water, extracted into EtOAc (2x), and the organic fractions were combined, dried (Na2SO4), and concentrated. The residue was purified by silica column chromatography (10-20% EtOAc/hexanes) to afford 59-C. LCMS-ESI+ (m/z): [M+H]+ calculated for C29H39NO5S1: 509.71; found: 510.793.
Step 3
A solution of 59-C (1.23 g, 2.41 mmol) in THF (13 ml) was stirred in an ice bath as 2.0 M L1BH4 in THF (2.42mL, 4.84 mmol)) was added and the resulting mixture was stirred at room température for 18 h. After the reaction mixture was diluted with ethyl acetate water was added slowly, two phases were separated, and the separated aqueous fraction was extracted with ethyl acetate. The two organic fractions were washed with water, combined, dried (Na2SÛ4), and concentrated. The residue was purified by silica column chromatography (20-40% EtOAc/hexanes) to afford 59-D. LCMS-ESI4 (m/z): [M+H]+ calculated for C28H39NO4S1: 481.7; found: 482.741.
Step 4
A mixture of 59-D (963 mg, 2.0 mmol), phthalimide (482 mg, 3.28 mmol) and triphenylphosphine (1.18 g, 4.49 mmol) in THF (50 ml) was cooled in an ice bath. Diisopropyl azodicarboxylate, 95% (0.89 mL, 4.52 mmol) was added. The mixture was then warmed up to room température and stirred for 20 h. After, the reaction mixture was concentrated and the residue dissolved in ether, cooled in an ice bath and stirred for 1.5 h. After, the solids were filtered off and the filtrate was concentrated. The residue was purified by silica column chromatography (10-30% EtOAc/hexanes) to afford the phthalimide adduct. LCMS-ESI+ (m/z): [M+H]+ calculated for C36H42N2O5S1: 610.81; found: 611.935.
A solution ofthe phthalimide adduct (1.2 g, 1.97 mmol) and hydrazine hydrate (0.4 ml, 8.03 mmol) in éthanol (12 ml) was stirred at 70 °C for 2h. The reaction mixture was cooled in an ice bath and ether (10 ml) was added, the mixture was stirred for 30 min. The solid formed was filtered off and the filtrate was concentrated under vacuum to dryness to afford 59-E. LCMS-ESI+ (m/z): [M+H]+ calculated for
132
C2sH4oN203Si.· 480.71; found: 481.356.
Step 5
A mixture of crude 59-E (770 mg, 1.60 mmol), compound 38-F (Example 38) (555 mg, 1.60 mmol) and NaHCO3 (269 mg, 3.20 mmol) in water (12 mL) and EtOH (12 mL) was stirred for 20 h. The reaction mixture was concentrated under vacuum and the residue was partitioned between water and EtOAc. The aqueous layer was reextracted with EtOAc and the combined organic layers were dried (Na2SÛ4) and concentrated.
The residue (1.29 g) was dissolved in CH2C12 (4 mL) and 4N HCl in dioxane (15.6 mL) was added. After 1.5 hours the mixture was concentrated to dryness, co-evaporated with toluene and dried in vacuum. LCMS-ESI+ (m/z): [M+H]+ calculated for C4iH48N2O7Si: 708.91; found: 709.782.
The crude residue (1.09 mg) and DBU (1.17 ml, 7.8 mmol) in toluene (20 ml) was stirred at 110 °C. After 35 min the mixture was concentrated and the residue was purified by silica column chromatography (5-15% MeOH/EtOAc) to afford 59-F. LCMS-ESI+ (m/z): [M+H]+ calculated for C39H42N2O6Si: 662.85; found: 663.677.
Step 6
A mixture of 59-F (680 mg, 1.03 mmol) in MeOH (10 ml) and THF (10 ml) was stirred at room température as IM KOH (5.42 ml) was added. After 30 min the reaction mixture was neutralized with IN HCl, extracted into EtOAc (2x) and the combined organic extracts were dried (Na2SO4) and concentrated. LCMS-ESI+ (m/z): [M+H]+ calculated for C37H38N20eSi: 634.79; found: 635.466.
A suspension of the crude residue (650 mg), 2,4,6-trifluorobenzylamine (214 mg, 1.33 mmol) and HATU (623 mg, 1.64 mmol) in dichloromethane (6 ml) was stirred at room température as Ν,Ν-diisopropylethylamine (DIPEA) (1.34 ml, 7.68 mmol) was added. After 2 h, the mixture was diluted with water, extracted into EtOAc (3x) nad the combined organic phases were dried (Na2SO4), concentrated and the residue was purified by silica column chromatography (50-75% EtOAc/hexanes) to afford 59-G. LCMS-ESI+ (m/z): [M+H]+ calculated for C44H42F3N3O5Si: 777.9; found: 778.566.
133
Step 7
A solution of 59-G (648 mg, 0.83 mmol ) in THF (10 ml) was stirred in an ice bath as 1.0 M tetrabutylammonium fluoride in THF (0.83 ml) was added dropwise and the resulting mixture was stirred at room température for 30 min. Additional 1.0 M tetrabutylammonium fluoride in THF (0.1 ml) was added dropwise. After 30 minutes, the reaction mixture was concentrated under vacuum and the residue was diluted with EtOAc, washed with water, dried (Na2SO4), concentrated and the residue was purified by silica column chromatography (5% MeOH/EtOAc). A solution of the residue (290 mg, 0.54 mmol) in dichloromethane (3 ml)was cooled to -78°C as diethylaminosulfur trifluoride (0.09 mL, 0.65 mmol) was added dropwise and the reaction was warmed to room température and stirred for 2.5 h. The reaction was cooled in an ice bath, quenched with saturated NaHCCb, two phases were separated, and the separated aqueous fraction was extracted with CH2CI2. The two organic fractions were combined, dried (Na2SO4), and concentrated. The residue was purified by silica column chromatography (1% MeOH/EtOAc) to afford 59-H. LCMS-ESI+ (m/z): [M+H]+ calculated for C28H23F4N3CU: 541.49; found: 542.320.
Step 8
Compound 59-H (103 mg, 0.19 mmol) was dissolved in TFA (1.4 mL) at room température for 15 min, and the solution was concentrated. The residue was suspended in DMF, filtered off, and the precipitated product was washed with water, dried under vacuum to afford compound 59. LCMS-ESI+ (m/z): [M+H]+ calculated for C2iHi7F4N3O4: 451.37, found: 452.226. ’H-NMR (400 MHz, DMSO-d6) δ 11.53 (s, IH), 10.35 (t, J = 5.8 Hz, IH), 8.34 (s, IH), 7.18 (t, J = 8.6 Hz, 2H), 5.15-4.88 (m, IH), 4.73 (d, J = 3.3 Hz, IH), 4.49 (m, 3H), 4.04 (t, J = 12.4 Hz, IH), 3.65 (dd, J - 12.4, 3.7 Hz, IH), 2.95-2.76(m, IH), 2.26-2.03 (m, IH), 1.96-1.64(m, 3H). 19F-NMR(376MHz, DMSO-i/e) δ -73.93 , -74.74 (d, J= 28.8 Hz), -109.31 (m), -112.51 (m), -165.65 (m).
Example 60
Préparation of Compound 60 (lR,4S,12aR)-N-(2,3-dichlorobenzyI)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9-carboxamide
134
H NH
41-E
O
MeO.
1) Π I 0
NaHCO3 0 OMe
2) HCl
3) NaHCO3
Step 1
To a solution of dimethyl 3-methoxy-4-oxo-4H-pyran-2,5-dicarboxylate (5.5 g, 23 mmol) in MeOH (100 mL) was added 41-E (Example 41) (5 g, 22 mmol) and sodium bicarbonate (3.6 g, 43 mmol). The solution was stirred at room température for
1.5 h. 4M HCl (in dioxane, 55 mL, 221 mmol) was added and the solution was heated to 50 °C for 2h. The reaction was cooled to room température and concentrated in vacuo. The resulting oil was dissolved in sodium bicarbonate and washed with EtOAc. The aqueous layers were then extracted with CH2CI2 (4x). The combined CH2Q2 extractions
135 were dried over NaiSCU and concentrated to provide 60-A. LCMS-ESE (m/z): [M+H]+ calculated for C16H19N2O5: 319.13; found: 319.20.
Step 2
To a suspension of 60-A (3.7 g, 11.6 mmol) in MeOH (12 mL) and THF (23 mL) was added aqueous KOH (2M, 15.7 mL, 31.4 mmol). The resulting solution was stirred at room température for 10 min. Volatiles were removed in vacuo, and the resulting aqueous layer was acidified with IN HCl. The resulting white solid was filtered, washed with water, and dried in vacuo to provide 60-B. ’H-NMR (400 MHz, Chloroform-d) δ 8.36 (s, IH), 5.01 (d, J = 2.7 Hz, IH), 4.12 (s, 4H), 3.90 (t, J = 12.2 Hz, IH), 3.78 (dd, J = 12.1, 3.1 Hz, IH), 2.69 (s, IH), 1.95 - 1.71 (m, 4H), 1.70 - 1.54 (m, 2H). LCMS-ESL (m/z): [M+H]+ calculated for C15H17N2O5: 305.11; found: 305.15.
Step 3
To a solution of 60-B (0.10 g, 0.33 mmol) in CH2CI2 (3.5 mL) was added (2,3-dichlorophenyl)methanamine (0.12 g, 0.70 mmol), HATU (0.25 g, 0.66 mmol), and Ν,Ν-diisopropylethylamine (DIPEA) (0.29 mL, 1.64 mmol). The resulting solution was stirred at room température until judged complété by LC/MS. The reaction mixture was diluted with CH2Q2 and washed with IN HCl. The aqueous layer was back-extracted with CH2CI2, and the combined organic layers were dried over Na2SC>4 and concentrated in vacuo. The crude material was dissolved in hot DMF and allowed to precipitate upon cooling. Filtration provided 60-C. LCMS-ESL (m/z): [M+H]+ calculated for C22H22CI2N3O4: 462.10; found: 462.14.
Step 4
To a slurry of 60-C (0.11 g, 0.24 mmol), in acetonitrile (4.5 mL), was added magnésium bromide (0.089 g, 0.48 mmol). The reaction mixture was heated to 45 °C for 2.5 h and then cooled to room température. The slurry was diluted with CH2CI2 and washed with IN HCl and brine. The aqueous layers were back-extracted with CH2CI2 (2x) and the combined organic layers were dried over NaiSCU and concentrated in vacuo. The crude solid was triturated with methanol and filtered to provide 60. 1H-NMR (400 MHz, DMSO-d6) δ 11.72 (s, IH), 10.50 (t, IH), 8.34 (s, IH), 7.55 (dd, IH), 7.40 - 7.24
136 (m, 2H), 4.67 (s, IH), 4.61 (d, 2H), 4.45 (dd, IH), 3.95 (t, IH), 3.84 - 3.73 (m, IH), 1.86
- 1.67 (m, 3 H), 1.66 - 1.40 (m, 4H). LCMS-ESF (m/z): [M+H]+ calculated for
C21H20CI2N3O4: 448.08; found: 448.18.
Example 61
Préparation of Compound 61 (lR,4S,12aS)-N-(3-chloro-2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9-carboxamide
O OH
H NH
2) HCl
3) NaHCO3
4) KOH
HATU, DIEA
F
137 was prepared analogously to Example 60, substituting (lS,3S,4R)-tertbutyl 3-(aminomethyl)-2-azabicyclo[2.2. i]heptane-2-carboxylate (prepared in Example 55) for 41-E, and (3-chloro-2,4-difluorophenyl)methanamine for (2,3dichlorophenyl)methanamine. ’H-NMR (400 MHz, DMSO-d6) δ 11.85 (s, IH), 10.45 (t, IH), 8.40 (s, IH), 7.37 (td, IH), 7.27 (td, IH), 4.63 - 4.46 (m, 4H), 4.17 (t, IH), 4.04 (dt, IH), 1.76 (d, IH), 1.73 - 1.54 (m, 5H). LCMS-ESH (m/z): [M+H]+ calculated for C21H19CIF2N3O4: 450.10; found: 450.15.
Example 62
Préparation of Compound 62 '(2R,5S,13aR)-N-(4-fluoro-2-(trifluoromethyl)benzyl)-8-hydroxy-7,9-dioxo2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb] [ 1,3 ]oxazepine-10-carboxamide
F
O OH
Compound 62 was prepared in a similar manner to compound 42 using (4fluoro-2-(trifluoromethyl)phenyl)methanamine in place of (2,4,6trifulorophenylphenyl)methanamine. 1H-NMR (400 MHz, Chloroform-d) δ 10.50 (s, IH), 8.38 (s, IH), 7.57 (dd, IH), 7.36 (dd, IH), 7.19 (td, IH), 5.40 - 5.28 (m, 2H), 4.79 (t, 2H), 4.69 (s, IH), 4.25 (dd, IH), 4.03 (dd, IH), 2.17 - 1.98 (m, 4H), 1.96 - 1.84 (m, IH), 1.61 (dt, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C22H20F4N3O5: 482.13; found: 482.145.
Example 63
Préparation of Compound 63 (2R, 5 S, 13 aR)-N-(2-chloro-4-fluorobenzyI)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13 aoctahydro-2,5 -methanopyrido[ 1 ',2' :4,5]pyrazino [2,1 -b] [ 1,3 Joxazepine-10-carboxamide
138
Compound 63 was prepared in a similar manner to compound 42 using (2chloro-4-fluorophenyl)methanamine in place of (2,4,6trifulorophenylphenyl)methanamine. 1H-NMR (400 MHz, Chloroform-d) δ 10.48 (s, IH), 8.45 (s, IH), 7.39 (dd, IH), 7.12 (dd, IH), 6.93 (td, IH), 5.37 (d, IH), 5.31 (t, IH), 4.68 (s, 3H), 4.29 (d, IH), 4.04 (t, IH), 2.21 - 2.01 (m, 4H), 1.97 - 1.82 (m, IH), 1.67 1.56 (m, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H20CIFN3O5: 448.10; found: 448.143.
Example 64
Préparation of Compound 64 (2R,5S,13aR)-8-hydroxy-7,9-dioxo-N-(2,4,5-trifluorobenzyl)-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[l',2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Compound 64 was prepared in a similar manner to compound 42 using (2,4,5-trifluorophenyl)methanamine in place of (2,4,6trifulorophenylphenyl)methanamine. *H-NMR (400 MHz, Chloroform-d) δ 10.42 (s, IH), 8.42 (s, IH), 7.19 (ddd, IH), 6.91 (td, IH), 5.38 (dd, IH), 5.31 (t, IH), 4.69 (s, IH), 4.61 (d, 2H), 4.29 (dd, IH), 4.05 (dd, IH), 2.18 - 2.02 (m, 4H), 1.96 - 1.84 (m, IH), 1.66 -1.56 (m, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H19F3N3O5: 450.12; found: 450.119.
139
Example 65
Préparation of Compound 65 (2R,5S,13aR)-N-(5-chloro-2,4-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13aoctahydro-2,5-methanopyrido[l',2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Compound 65 was prepared in a similar manner to compound 42 using (5chloro-2,4-difluorophenyl)methanamine in place of (2,4,6trifùlorophenylphenyl)methanamine. ’H-NMR (400 MHz, Chloroform-d) δ 10.47 (t, IH), 8.41 (s, IH), 7.40 (dd, IH), 6.90 (t, IH), 5.37 (dd, IH), 5.31 (t, IH), 4.69 (s, IH), 4.62 (d, 2H), 4.28 (d, IH), 4.04 (dd, IH), 2.17 - 2.02 (m, 4H), 1.94 - 1.86 (m, IH), 1.61 (dt, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H19CIF2N3O5: 466.09; found: 466.107.
Example 66
Préparation of Compound 66 (lR,4S,12aR)-N-(3,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
Compound 66 was prepared in a similar manner to compound 60 using (3,4-difluorophenyl)methanamine in place of (2,3-dichlorophenyl)methanamine. ΉNMR (400 MHz, Chloroform-d) δ 10.59 (s, IH), 7.24 - 7.16 (m, 2H), 7.14 - 7.04 (m,
140
2H), 4.91 (s, IH), 4.58 (d, 3H), 3.94 - 3.82 (m, IH), 3.79 (d, IH), 1.99 - 1.81 (m, 4H),
1.76 (d, IH), 1.70 - 1.60 (m, 3H). LCMS-ESF (m/z): [M+H]+ calculated for C21H20F2N3O4: 416.13; found: 416.415.
Example 67
Préparation of Compound 67 (lR,4S,12aR)-N-(4-fluoro-2-(trifluoromethyl)benzyl)-7-hydroxy-6,8-dioxol,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9carboxamide
Compound 67 was prepared in a similar manner to compound 60 using (4fluoro-2-(trifluoromethyl)phenyl)methanamine in place of (2,3dichlorophenyl)methanamine. ’H-NMR (400 MHz, Chloroform-ûQ δ 11.72 (s, IH), 10.55 (s, IH), 8.29 (s, IH), 7.61 (s, IH), 7.36 (dd, IH), 7.18 (td, IH), 4.91 (s, IH), 4.80 (d, 3H), 4.11 (s, IH), 1.99 - 1.80 (m, 4H), 1.76 (d, IH), 1.71 - 1.47 (m, 3H). LCMSESF (m/z): [M+H]+ calculated for C22H20F4N3O4: 466.13; found: 466.297.
Example 68
Préparation of Compound 68 ( 1 R,4S, 12aR)-N-(2-chloro-4-fluorobenzyl)-7-hydroxy-6,8-dioxo- 1,2,3,4,6,8,12,12aoctahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
141
Compound 68 was prepared in a similar manner to compound 60 using (2chloro-4-fluorophenyl)methanamine in place of (2,3-dichlorophenyl)methanamine. *ΗNMR (400 MHz, Chloroform-ûQ δ 11.68 (s, IH), 10.52 (s, IH), 8.27 (s, IH), 7.44 - 7.37 (m, IH), 7.11 (dd, IH), 6.93 (td, IH), 4.90 (s, IH), 4.68 (d, 2H), 4.16 - 4.01 (m, IH), 3.88 - 3.70 (m, 2H), 2.00 - 1.79 (m, 4H), 1.75 (d, IH), 1.70 - 1.57 (m, 2H). LCMSESI+ (m/z): [M+H]+ calculated for C21H20CIFN3O4: 432.10; found: 432.214.
Example 69
Préparation of Compound 69 (1R,4S, 12aR)-N-(3-chloro-2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9-carboxamide
Compound 69 was prepared in a similar manner to compound 60 using (3chloro-2,4-difluorophenyl)methanamine in place of (2,3-dichlorophenyl)methanamine. ’H-NMR (400 MHz, Chloroformé δ 11.71 (s, IH), 10.48 (s, IH), 8.26 (s, IH), 7.27 (s, IH), 6.92 (td, IH), 4.90 (s, IH), 4.66 (d, 2H), 4.08 (s, IH), 3.91 - 3.69 (m, 2H), 2.01 1.79 (m, 3H), 1.75 (d, IH), 1.71 - 1.44 (m, 2H). LCMS-ESF (m/z): [M+H]+ calculated for C21H19CIF2N3O4: 450.10; found: 450.27.
Example 70
Préparation of Compound 70 (1R,4S, 12aR)-N-(2-fluoro-3-methylbenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9-carboxamide
142
Compound 70 was prepared in a similar manner to compound 60 using (2fluoro-3-methylphenyl)methanamine in place of (2,3-dichlorophenyl)methanamine. 1HNMR (400 MHz, Chloroform-d) δ 11.62 (s, IH), 10.39 (s, IH), 8.30 (s, IH), 7.19 (t, IH), 7.07 (t, IH), 6.96 (t, IH), 4.89 (d, IH), 4.67 (d, 2H), 4.08 (s, IH), 3.88 - 3.67 (m, 2H), 2.26 (d, 3H), 1.97 - 1.79 (m, 3H), 1.78 - 1.39 (m, 3H). LCMS-ESU (m/z): [M+H]+ calculated for C22H23FN3O4: 412.16; found: 412.26.
Example 71
Préparation of Compound 71 (lR,4S,12aR)-N-(3,6-dichloro-2-fluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
Compound 71 was prepared in a similar manner to compound 60 using (3,6-dichloro-2-fluorophenyl)methanamine in place of (2,3dichlorophenyl)methanamine. IH-NMR (400 MHz, Chloroform-d) δ 11.62 (s, IH), 10.47 (t, IH), 8.29 (s, IH), 7.13 (dd, IH), 4.88 (s, IH), 4.85 - 4.73 (m, 2H), 4.09 (d, IH), 3.88 - 3.68 (m, 2H), 1.99 - 1.53 (m, 8H). LCMS-ESE (m/z): [M+H]+ calculated for C21H19CI2FN3O4: 466.07; found: 466.257.
143
Example 72
Préparation of Compound 72 (lR,4S,12aR)-N-(3-chlorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12a-octahydro1,4-methanodipyrido[ 1,2-a: l',2'-d]pyrazine-9-carboxamide
Compound 72 was prepared in a similar manner to compound 60 using (3chlorophenyl)methanamine in place of (2,3-dichlorophenyl)methanamine. ’H-NMR (400 MHz, DMSO-d6) Ô 11.75 (s, IH), 10.44 (t, IH), 8.38 (s, IH), 7.42 - 7.22 (m, 4H),
4.68 (s, IH), 4.54 (d, 2H), 4.48 (dd, IH), 3.97 (t, IH), 3.81 (dd, IH), 2.58 (s, IH), 1.87 -
1.69 (m, 3H), 1.68 - 1.51 (m, 2H), 1.46 (d, IH). LCMS-ESF (m/z): [M+H]+ calculated for C21H21CIN3O4: 414.11; found: 414.21.
Example 73
Préparation of Compound 73 (lR,4S,12aR)-N-(3-chloro-2,6-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
Compound 73 was prepared in a similar manner to compound 60 using (3chloro-2,6-difluorophenyl)methanamine in place of (2,3-dichlorophenyl)methanamine. 1H-NMR (400 MHz, DMSO-d6) δ 11.71 (s, IH), 10.46 (t, IH), 8.34 (s, IH), 7.60 (td, IH), 7.19 (td, IH), 4.67 (s, IH), 4.62 (d, 2H), 4.44 (dd, IH), 3.95 (t, IH), 3.78 (dd, IH),
144
2.57 (s, IH), 1.86 - 1.68 (m, 3H), 1.67 - 1.49 (m, 2H), 1.45 (d, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H19CIF2N3O4: 450.10; found: 450.16.
Example 74
Préparation of Compound 74 (lR,4S,12aR)-N-(2-fluoro-3-(trifluoromethyl)benzyl)-7-hydroxy-6,8-dioxo1,2,3,4,6,8,12,12a-octahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9carboxamide
Compound 74 was prepared in a similar manner to compound 60 using (2fluoro-3-(trifluoromethyl)phenyl)methanamine in place of (2,3dichlorophenyl)methanamine. ‘H-NMR (400 MHz, DMSO-d6) δ 11.76 (s, IH), 10.48 (t, IH), 8.36 (s, IH), 7.68 (q, 2H), 7.38 (t, IH), 4.68 (s, IH), 4.65 (d, 2H), 4.47 (dd, IH), 3.96 (t, IH), 3.80 (dd, IH), 2.57 (s, IH), 1.88 - 1.69 (m, 3H), 1.67 - 1.50 (m, 2H), 1.45 (d, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C22H20F4N3O4: 466.13; found: 466.142.
Example 75
Préparation of Compound 75 (1R,4S, 12aR)-N-(3-chloro-4-fluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9-carboxamide
145
Compound 75 was prepared in a similar manner to compound 60 using (3chloro-4-fluorophenyl)methanamine in place of (2,3-dichlorophenyl)methanamine. ’HNMR (400 MHz, DMSO-d6) δ 11.75 (s, IH), 10.43 (t, IH), 8.38 (s, IH), 7.51 (dd, IH), 7.42 - 7.28 (m, 2H), 4.68 (s, IH), 4.51 (d, 2H), 4.47 (dd, IH), 3.97 (t, IH), 3.80 (dd, IH), 2.58 (s, IH), 1.86 - 1.68 (m, 3H), 1.68 - 1.52 (m, 2H), 1.46 (d, IH). LCMS-ESI4 (m/z): [M+H]+ calculated for C21H20CIFN3O4: 432.10; found: 432.159.
Example 76
Préparation of Compound 76 (lR,4S,12aR)-N-((3,5-difluoropyridin-2-yl)methyl)-7-hydroxy-6,8-dioxo1,2,3,4,6,8,12,12a-octahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9-
Compound 76 was prepared in a similar manner to compound 60 using (3,5-difluoropyridin-2-yl)methanamine in place of (2,3-dichlorophenyl)methanamine. 1H-NMR (400 MHz, Chloroform-d) δ 10.80 (s, IH), 8.81 (s, IH), 8.33 (d, IH), 7.20 (td, IH), 4.90 (s, IH), 4.82 (s, 2H), 4.28 (d, IH), 3.92 - 3.75 (m, 2H), 3.48 (s, 2H), 1.98 1.80 (m, 3H), 1.77 (d, IH), 1.71 - 1.58 (m, 2H). LCMS-ESE (m/z): [M+H]+ calculated for C20H19F2N4O4: 417.13; found: 417.189.
Exampie 77
Préparation of Compound 77 ( 1R,4S, 12aR)-7-hydroxy-6,8-dioxo-N-((R)-1 -(2,4,6-trifluorophenyl)ethyl)1,2,3,4,6,8,12,12a-octahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9carboxamide
146
Step 1
A 50-mL round bottom flask was charged with 77-A (0.15 g, 0.39 mmol), (7?)-l-(2,4,6-trifluorophenyl)ethanamine (0.14 g, 0.78 mmol), N,Ndiisopropylethylamine (DIPEA) (0.25 g, 1.97 mmol) and HATU (0.29 g, 0.79 mmol) in DCM (10 ml). The reaction mixture was stirred at room température for 1 h. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 mL), washed with saturated NaHCO3 (2x), saturated NH4CI and dried over Na2SO4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 77B as a white solid. LCMS-ESU (m/z): [M+H]+ found: 538.
Step 2
A 50-mL round bottom flask was charged with 77-B (0.20 g, 0.37 mmol) in TFA (2 mL). The reaction mixture was stirred at room température for 30 min. The solution was concentrated and the residue was purified by flash chromatography using EtOAc-20% MeOH in EtOAc as eluents to afford compound 77. 1H-NMR (400 MHz, Chloroform-d) δ 10.67 (d, J = 8.2 Hz, IH), 8.22 (s, IH), 6.61 (t, J = 8.4 Hz, 2H), 5.60 (dd, J = 8.1, 6.9 Hz, IH), 4.85 (s, IH), 3.82 (t, J = 12.2 Hz, IH), 3.71 (dd, J = 12.4, 3.4 Hz, IH), 2.75 - 2.55 (m, 3H), 1.97 - 1.57 (m, 9H). 19F-NMR (376 MHz, Chloroform-d)
147 δ -109.65 - -111.29 (m), -111.76 --113.09 (m) LCMS-ESF (m/z): [M+Hf found: 448.
Example 78
Préparation of Compound 78 (2R, 13 aR)-8-hydroxy-7,9-dioxo-N-((R)-1 -(2,4,6-trifluorophenyl)ethyl)2,3,4,5,7,9,13,13a-octahydro-2,5-methanopyrido[r,2':4,5]pyrazino[2,lb] [ 1,3 ]oxazepine-10-carboxamide
Step 1
A 50-mL round bottom flask was charged with 78-A (0.30 g, 0.94 mmol), (R)-l-(2,4,6-trifluorophenyl) ethanamine (0.39 g, 1.87 mmol), N,Ndiisopropylethylamine (DIPEA) (0.61 g, 4.87 mmol) and HATU (0.71 g, 1.87 mmol) in DCM (10 ml). The reaction mixture was stirred at room température for 1 hour. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 mL), washed with saturated NaHCO3 (2x), saturated NH4C1 and dried over Na2SO4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 78-B as a white solid. LCMS-ESI^ (rn/z): [M+H]+; found: 478.
148
Step 2
A 50-mL round bottom fiask was charged with 78-B (0.4 g, 0.84 mmol) and magnésium bromide (0.4 g, 2.2 mmol) in acetonitrile (5 mL). The reaction mixture was heated to 50 °C. After 10 minutes, the reaction mixture was cooled to 0 °C and 1 N hydrochloric acid (4 mL) was added in. More water (~ 5 mL) was added and the solid was filtrated and washed with water and dried to afford afford compound 78. ’H-NMR (400 MHz, Chloroform-d) δ 12.30 (s, IH), 10.59 (d, J = 8.3 Hz, IH), 8.21 (s, IH), 6.60 (t, J = 8.4 Hz, 2H), 5.59 (t, J = 7.4 Hz, IH), 5.37 (dd, J = 9.4, 4.1 Hz, IH), 5.31-5.09 (m, IH), 4.64 (t, J = 3.0 Hz, IH), 4.20 (dd, J = 12.9, 4.1 Hz, 2H), 3.96 (dd, J = 12.8, 9.4 Hz, 2H), 2.21 - 1.85 (m, 4H), 1.71 - 1.43 (m, 3H). 19F-NMR(376 MHz, Chloroform-d) δ 110.37 (tt, J = 8.7, 6.1 Hz), -112.19 (t, J = 7.2 Hz). LCMS-ESF (m/z): [M+H]+ found: 464.
Example 79
Préparation of Compound 79 (lR,4S,12aR)-7-hydroxy-6,8-dioxo-N-(2,4,5-trifluorobenzyl)-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9-carboxamide
149
Step 1
A 50-mL round bottom flask was charged with 79-A (0.12 g, 0.32 mmol), (2,4,5-trifluorophenyl)methanamine (0.10 g, 0.63 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.20 g, 1.58 mmol) and HATU (0.24 g, 0.63 mmol) in DCM (10 ml). The reaction mixture was stirred at room température for 1 h. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 mL), washed with saturated NaHCÛ3 (2x), saturated NH4CI and dried over Na2SÛ4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 79-B as a white solid. LCMS-ESI+ (m/z): [M+H]+; found: 524.
Step 2
A 50-mL round bottom flask was charged with 79-B (0.15 g, 0.29 mmol) in TFA (2 mL). The reaction mixture was stirred at room température for 30 min. The solution was concentrated and the residue was purified by flash chromatography using EtOAc-20% MeOH in EtOAc as eluents to afford compound 79. 1H-NMR (400 MHz, Chloroform-d) δ 11.70 (s, IH), 10.65-10.18 (m, IH), 8.27 (s, IH), 7.26 (m, IH), 6.90 (td, J = 9.7, 6.4 Hz, IH), 4.89 (s, IH), 4.60 (d, J = 6.0 Hz, 2H), 4.09 (dd, J = 11.4, 2.6 Hz, IH), 3.96 - 3.66 (m, 2H), 2.68 (s, IH), 2.15 - 1.43 (m, 6H). 19F-NMR (376 MHz, Chloroform-d) δ 120.53 - -120.85 (m), -134.68 - -136.79 (m), -142.26 - -144.11 (m). LCMS-ESI+ (m/z): [M+H]+ found: 434.
150
Example 80
Préparation of Compound 80 (lR,4S,12aR)-N-(5-chloro-2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9-carboxamide
Step 1
A 50-mL round bottom flask was charged with 80-A (0.12 g, 0.32 mmol), (5-chloro-2,4-difluorophenyl)methanamine (0.11 g, 0.63 mmol), N,Ndiisopropylethylamine (DIPEA) (0.20 g, 1.58 mmol) and HATU (0.24 g, 0.63 mmol) in DCM (10 ml). The reaction mixture was stirred at room température for 1 h. The reaction mixture was concentrated down, re-dissolved in EtOAc (50 mL), washed with saturated NaHCO3 (2x), saturated NH4Ci and dried over Na2SÛ4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to obtain 80B as a white solid. LCMS-ESF (m/z): [M+H]+; found: 541.
Step 2
A 50-mL round bottom flask was charged with 80-B (0.14 g, 0.26 mmol) in TFA (2 mL). The reaction mixture was stirred at room température for 30 minutes. The solution was concentrated and the residue was purified by flash chromatography using
151
EtOAc-20% MeOH in EtOAc as eluents to afford compound 80. 1H-NMR (400 MHz, Chloroform-d) δ 10.46 (s, IH), 8.27 (s, IH), 7.40 (t, J = 7.8 Hz, IH), 6.89 (t, J = 9.1 Hz, IH), 4.90 (s, IH), 4.78 - 4.48 (m, 2H), 4.08 (dd, J = 11.3, 2.5 Hz, IH), 3.95 - 3.63 (m, 2H), 2.68 (s, IH), 2.22 - 1.51 (m, 7H). 19F-NMR (376 MHz, Chloroform-d) δ -113.37 5 (q, J = 8.1 Hz), -116.37 (q, J = 8.0 Hz). LCMS-ESE (m/z): [M+H]+ found: 451.
Example 81
Préparation of Compound 81 (lR,3S,4S,12aS)-3-fluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)10 l,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9 carboxamide
152
DAST/DCM
Hydrazine
l2O
) HCI/dioxane
2) DBU/Tol
Step 1
A 100-mL round bottom flask was charged with 81-A (1.0 g, 3.7 mmol) in DCM (10 mL). The reaction mixture was cooled to 0 °C. Diethylaminosulfur trifluoride (DAST) (0.58 mL, 4.1 mmol) was slowly added in. Then the reaction mixture was stirred at room température for one hour. The mixture was cooled back to 0 °C. Saturated NaHCCh (5 mL) was added dropwise to quench the reaction. Then the reaction mixture was diluted with EtOAc (100 mL), washed with sat. NaHCÛ3, brine, and dried over
153
Na2SO4. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 81-B. LCMS-ESC (m/z): [M+H]+ found: 274.
Step 2
A 100-mL round bottom flask was charged with 81-B (0.8 g, 3.0 mmol) in THF (10 mL). The reaction mixture was stirred at -78 °C. 2.0 M LiBH4 in THF (3.2 mL, 6.4 mmol) was slowly added in. Then the reaction mixture was warmed up and stirred at room température for 3 hours. Then the reaction mixture was diluted with EtOAc (100 mL) and treated slowly with water (H2 évolution). After the two phases were separated, the aqueous fraction was extracted with EtOAc and the two organic fractions were combined, washed with water, and dried over Na2SO4. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 81-C. LCMS-ESI+ (m/z): [M+H]+ found: 246.
Step 3
A 100-mL round bottom flask was charged with 81-C (0.57 g, 2.3 mmol), triphenylphosphine (1.3 g, 5.1 mmol) and phthalimide (0.55 g, 3.7 mmol) in THF (15 mL). Then the reaction mixture was cooled to 0 °C with stirring. Diisopropyl azodicarboxylate (DIAD) (1.0 mL, 5.1 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room température for ovemight. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 81-D. LCMS-ESI+ (m/z): [M+H]+ found: 375.
Step 4
To a solution of 81-D (0.8 g, 2.1 mmol) EtOH (40 mL) was added hydrazine monohydrate (0.6 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 81E. LCMS-ESI+ (m/z): [M+H]+found: 245.
Step 5
A 100-mL round bottom flask was charged with 81-E (0.49 g, 2.0 mmol) and 81-F (0.7 g, 2.0 mmol) in éthanol (7 mL). Sodium bicarbonate (0.34 g, 4.0 mmol) in
154 water (7 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for ovemight. The mixture was diluted with EtOAc (50 mL) and washed with water (2 x). The aqueous fractions were extracted with EtOAc (1 x), and the organic fractions were combined, dried (Na2SO4), and concentrated. The crude 81G was used for next step without further purification. LCMS-ESI+ (m/z): [M+H]+ found: 573.
Step 6
A 100-mL round bottom flask was charged with 81-G (1.1 g, 1.9 mmol) in 4 N HCl /dioxane (11 mL). Then the reaction mixture was stirred at room température for 1 hour. After concentration, 1.0 g intermediate was obtained. The intermediate and DBU (1.3 g, 8.8 mmol) were dissolved in toluene (10 mL). The reaction mixture was heated to 110 °C with stirring for 1 hour. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 81-H. LCMS-ESI+ (m/z): [M+H]+found: 413.
Step 7
A 100-mL round bottom flask was charged with 81-H (0.56 g, 1.4 mmol) in THF (5 mL) and MeOH (5 mL). 1 N KOH (4 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for 1 hour. The reaction mixture was acidified by adding 1 N HCl (4 mL). After concentration, the residue was co-evaporated with toluene (3 x). Half of the crude acid, 2,4,6-trifluobenzylamine (0.2 g, 1.3 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.41 g, 3.1 mmol) and HATU (0.48 g, 1.25 mmol) were dissolved in DMF (10 mL). The reaction mixture was stirred at room température for 2 hours. The mixture was diluted with EtOAc (100 mL) and washed with saturated NaHCOs (2x), saturated NH4CI (2x) and dried over Na2SO4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 81-1. LCMS-EST (m/z): [M+H]+found: 542.
Step 8
A 50-mL round bottom flask was charged with 81-1 (0.31 g, 0.58 mmol) in TFA (3 mL). The reaction mixture was stirred at room température for 30 minutes.
155
After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 81. 1H-NMR (400 MHz, Chloroform-d) δ 10.29 (s,
IH), 8.31 (s, IH), 6.65 (dd, J = 8.7, 7.5 Hz, 2H), 5.05 - 4.75 (m, 2H), 4.65 (d, J = 5.6 Hz, 2H), 4.11 (d, J= 12.2 Hz, IH), 3.83 (t, J = 12.3 Hz, IH), 3.56 (dd, J = 12.3, 3.3 Hz, IH),
2.77 (s, IH), 2.25 - 1.97 (m, 2H), 1.95 (d, J = 11.0 Hz, 2H), 1.77 (d, J= 11.2 Hz, IH).
19F-NMR (376 MHz, Chloroform-d) δ -108.98 (t, J = 8.2 Hz), -112.03 (t, J = 7.2 Hz), 168.00. LCMS-ESI+ (m/z): found: 452.
Example 82
Préparation of Compound 82 (1 S,3R,4R, 12aR)-3-fluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)1,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a: l',2'-d]pyr azine-9carboxamide
156
DAST/DCM
LîBH4/THF
Hydrazine
Step 1
A 100-mL round bottom flask was charged with 82-A (0.6 g, 2.1 mmol) 5 in DCM (6 mL). The reaction mixture was cooled to 0 °C. DAST (0.35 mL, 3.0 mmol) was slowly added in. Then the reaction mixture was stirred at room température for one hour. The mixture was cooled back to 0 °C. Saturated NaHCCh (5 mL) was added drop wise to quench the reaction. Then the reaction mixture was diluted with EtOAc (100 mL), washed with sat. NaHCÛ3, brine, and dried over Na2SÛ4. After concentration, the 10 residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford
157
82-B LCMS-EST (nvz): [M+H]* found: 274.
Step 2
A 100-mL round bottom flask was charged with 82-B (0.4 g, 1.5 mmol) in THF (10 mL). The reaction mixture was stirred at -78 °C. 2.0 M LiBH4 in THF (1.6 mL, 3.2 mmol) was slowly added in. Then the reaction mixture was warmed up and stirred at room température for 3 hours. Then the reaction mixture was diluted with EtOAc (100 mL) and added water slowly (H2 évolution). After the two phases were separated, the aqueous fraction was extracted with EtOAc and the two organic fractions were combined, washed with water and dried over Na2SO4. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 82-C. LCMS-ESU (m/z): [M+H]+ found: 246.
Step 3
A 100-mL round bottom flask was charged with 82-C (0.25 g, 1.0 mmol), triphenylphosphine (0.59 g, 2.2 mmol) and phthalimide (0.24 g, 1.6 mmol) in THF (10 mL). Then the reaction mixture was cooled to 0 °C with stirring. DIAD (0.44 mL, 2.2 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room température for ovemight. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 82-D. LCMS-ESI+ (m/z): [M+H]+ found: 375.
Step 4
To a solution of 82-D (0.35 g, 0.9 mmol) EtOH (20 mL) was added hydrazine monohydrate (0.3 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 82E. LCMS-ESI+ (m/z): [M+H]+ found: 245.
Step 5
A 100-mL round bottom flask was charged with 82-E (0.21 g, 0.87 mmol) and 82-F (0.3 g, 0.87 mmol) in éthanol (7 mL). Sodium bicarbonate (0.15 g, 1.7 mmol) in water (7 mL) was added to the reaction mixture. Then the reaction mixture was stirred
158 at room température for ovemight. The mixture was diluted with EtOAc (50 mL) and washed with water (2 x). The aqueous fractions were extracted with EtOAc, and the organic fractions were combined, dried (Na2SÜ4), and concentrated. The crude 82-G was used for next step without further purification. LCMS-ESI+ (m/z): [M+H]+found: 573.
Step 6
A 100-mL round bottom flask was charged with 82-G (0.49 g, 0.86 mmol) in 4 N HCl /dioxane (5 mL). Then the reaction mixture was stirred at room température for 1 hour. After concentration, 0.4 g intermediate was obtained. The intermediate and DBU (0.6 g, 4.0 mmol) were dissolved in toluene (10 mL). The reaction mixture was heated to 110 °C with stirring for 1 hour. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 82-H. LCMS-ESI+ (m/z): [M+H]+found: 413.
Step 7
A 100-mL round bottom flask was charged with 82-H (0.2 g, 0.49 mmol) in THF (5 mL) and MeOH (5 mL). 1 N KOH (1.5 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for 1 hour. The reaction mixture was acidified by adding 1 N HCl (1.5 mL). After concentration, the residue was co-evaporated with toluene (3 x). The crude acid, 2,4,6-trifluobenzylamine (0.15 g, 0.95 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.31 g, 2.4 mmol) and HATU (0.36 g, 0.95 mmol) were dissolved in DCM (10 mL). The reaction mixture was stirred at room température for 2 hours. The mixture was diluted with EtOAc (100 mL) and washed with saturated NaHCO3 (2x), saturated bffikCl (2x) and dried over Na2SO4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 82-1. LCMS-ESI+ (m/z): [M+H]+ found: 542.
Step 8
A 50-mL round bottom flask was charged with 82-1 (0.22 g, 0.41 mmol) in TFA (3 mL). The reaction mixture was stirred at room température for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 82. ’H-NMR (400 MHz, Chloroform-d) δ 10.25 (s,
159
IH), 8.28 (s, IH), 6.65 (s, 2H), 5.15 - 4.77 (m, 2H), 4.65 (s, 2H), 4.32 - 3.41 (m, 2H), 2.78 (s, IH), 1.86 (dd, J = 144.8, 72.3 Hz, 6H). 19F-NMR (376 MHz, Chloroform-d) δ 108.98 (t, J = 8.2 Hz), -112.03 (t, J = 7.2 Hz), -168.00. LCMS-ESU (m/z): found: 452.
Example 83
Préparation of Compound 83 (lS,4R,12aS)-3,3-difluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)l,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9carboxamide
160 ο
Ο
83-Β
Ο
83-C
83-Α
NaHCO3
EtOH/H2O
83-G
83-J
Step 1
A 100-mL round bottom flask was charged with 83-A (1.0 g, 3.7 mmol) in DCM (20 mL). The reaction mixture was cooled to 0 °C. Dess-Martin periodinane (1.8 g, 4.2 mmol) was slowly added in. Then the reaction mixture was stirred at room température for 3 hours. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-B. LCMS-ESU (m/z): [M+H]+ found: 270.
161
Step 2
A 100-mL round bottom flask was charged with 83-B (0.85 g, 3.2 mmol) in DCM (15 mL). The reaction mixture was cooled to 0 °C. DAST (1.5 mL, 11.3 mmol) was slowly added in. Then the reaction mixture was stirred at room température ovemight. The mixture was cooled back to 0 °C. Saturated NaHCCh (5 mL) was added dropwise to quench the reaction. Then the reaction mixture was diluted with EtOAc (100 mL), washed with sat. NaHCO3, brine, and dried over Na2SÛ4. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-C. LCMS-ESI+ (m/z): [M+H]+ found: 292.
Step 3
A 100-mL round bottom flask was charged with 83-C (0.44 g, 1.5 mmol) in THF (6 mL). The reaction mixture was stirred at -78 °C. 2.0 M LiBH4 in THF (1.6 mL, 3.2 mmol) was slowly added in. Then the reaction mixture was warmed up and stirred at room température for 3 hours. Then the reaction mixture was diluted with EtOAc (100 mL) and added water slowly (H2 évolution). After the two phases were separated, the aqueous fraction was extracted with EtOAc and the two organic fractions were combined, washed with water, and dried over Na2SO4. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-D LCMS-ESI+ (m/z): [M+H]+ found: 264.
Step 4
A 100-mL round bottom flask was charged with 83-D (0.17 g, 0.65 mmol), triphenylphosphine (0.37 g, 1.4 mmol) and phthalimide (0.15 g, 1.0 mmol) in THF (10 mL). Then the reaction mixture was cooled to 0 °C with stirring. DIAD (0.28 mL, 1.4 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room température for ovemight. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-E. LCMS-ESI4 (m/z): [M+H]+ found: 393.
162
Step 5
To a solution of 83-E (0.25 g, 0.64 mmol) EtOH (20 mL) was added hydrazine monohydrate (0.3 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 83F. LCMS-ESI+ (m/z): [M+H]+ found: 263.
Step 6
A 100-mL round bottom flask was charged with 83-F (0.18 g, 0.69 mmol) and 83-G (0.324g, 0.69 mmol) in éthanol (7 mL). Sodium bicarbonate (0.12 g, 1.4 mmol) in water (7 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température ovemight. The mixture was diluted with EtOAc (50 mL) and washed with water. The aqueous fractions were extracted with EtOAc, and the organic fractions were combined, dried (Na2SÛ4), and concentrated. The crude 83-H was used for next step without further purification. LCMS-ESI4 (m/z): [M+H]+found: 591.
Step 7
A 100-mL round bottom flask was charged with 83-H (0.4 g, 0.68 mmol) in 4 N HCl /dioxane (3.8 mL). Then the reaction mixture was stirred at room température for 1 hour. After concentration, 0.35 g intermediate was obtained. The intermediate and DBU (0.51 g, 3.3 mmol) were dissolved in toluene (10 mL). The reaction mixture was heated to 110 °C with stirring for 1 hour. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 83-1. LCMS-ESI+ (m/z): [M+H]+found: 431.
Step 8
A 100-mL round bottom flask was charged with 83-1 (0.2 g, 0.47 mmol) in THF (5 mL) and MeOH (5 mL). 1 N KOH (1.4 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for 1 hour. The reaction mixture was acidified by adding 1 N HCl (1.4 mL). After concentration, the residue was co-evaporated with toluene (3 x). The crude acid, 2,4,6-trifluobenzylamine (0.14 g, 0.91 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.29 g, 2.2 mmol) and HATU (0.35 g, 0.91 mmol) were dissolved in DCM (10 mL). The reaction mixture was stirred at room
163 température for 2 hours. The mixture was diluted with EtOAc (100 mL) and washed with saturated NaHCO3 (2x), saturated NH4CI (2x) and dried over Na2SO4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 83-J LCMS-ESI+ (m/z): [M+H]+ found: 560.
Step 9
A 50-mL rbf was charged with 83-J (0.18 g, 0.32 mmol) in TFA (3 mL).
The reaction mixture was stirred at room température for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 83 as a white solid. 1H-NMR (400 MHz, Chloroform-d) δ 10.29 (d, J = 6.1 Hz, IH), 8.34 (s, IH), 6.65 (dd, J = 8.7, 7.5 Hz, 2H), 4.83 (s, IH), 4.72 - 4.58 (m, 2H), 4.36 - 4.10 (m, 2H), 4.05 (t, J = 11.5 Hz, IH), 2.97 (d, J = 4.4 Hz, IH), 2.49 - 2.08 (m, 3H), 2.12 - 1.94 (m, 2H). 19F-NMR (376 MHz, Chloroform-d) δ - 92.32 (ddd, J = 225.6, 22.5, 9.1 Hz), -107.64 - -109.54 (m), -112.05 (t, J = 7.0 Hz), -114.67 (d, J = 226.7 Hz). LCMS-ESI+ (m/z): found: 470.
Example 84
Préparation of Compound 84 (lS,2R,4S,12aR)-7-hydroxy-2-methyl-6,8-dioxo-N-(2,4,6-trifluorobenzyl)l,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:l',2'-d]pyrazine-9carboxamide
O OH
164
DMP
84-B
Tebbe reagent
UBH4/THF
Hydrazine
84-G
84-H
1) HCI/dioxane
2) DBU/ToJ
84-I
TFA
Step 1
A 100-mL round bottom flask was charged with 84-A (1.6 g, 5.9 mmol) in DCM (20 mL). The reaction mixture was cooled to 0 °C. Dess-Martin periodinane (4.9 g, 11.7 mmol) was slowly added in. Then the reaction mixture was stirred at room
165 température for 3 hours. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 84-B. LCMS-ESI4 (m/z): [M+H]+ found: 270.
Step 2
A 100-mL round bottom flask was charged with 84-B (1.3 g, 4.8 mmol) in THF (30 mL). The reaction mixture was cooled to 0 °C. Tebbe reagent (0.5 M in toluene, 19.4 mL, 9.7 mmol) was slowly added in. Then the reaction mixture was stirred at room température for 2 hours. The mixture was cooled back to 0 °C. Saturated NaHCOs (5 mL) was added drop wise to quench the reaction. The reaction mixture was stirred at room température for another 15 minutes and fîltered through celite. The fîltered cake was washed with DCM (2 x). The combined filtrâtes were concentrated in vacuum and the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 84-C. LCMS-ESI+ (m/z): [M+H]+ found: 268.
Step 3
To a solution (purged with N2) of 84-C (0.9 g, 3.4 mmol) in EtOH (20 mL) was added Pd/C (0.18 g). The mixture was stirred under H2 for 3 hours. The mixture was fîltered through celite and the fîltrate was concentrated to afford 84-D. LCMS-ESI4 (m/z): [M+H]+found: 270.
Step 4
A 100-mL round bottom flask was charged with 84-D (0.9 g, 3.3 mmol) in THF (6 mL). The reaction mixture was stirred at -78 °C. 2.0 M L1BH4 in THF (13.2 mL, 26.4 mmol) was slowly added in. Then the reaction mixture was warmed up and stirred at room température for 3 hours. Then the reaction mixture was diluted with EtOAc (100 mL) and added water slowly (H2 évolution). After the two phases were separated, the aqueous fraction was extracted with EtOAc and the two organic fractions were combined, washed with water, and dried over Na2SO4. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 84-E LCMS-ESI+ (m/z): [M+H]+ found: 242.
166
Step 5
A 100-mL round bottom flask was charged with 84-E (0.4 g, 1.66 mmol), triphenylphosphine (0.96 g, 3.6 mmol) and phthalimide (0.39 g, 2.7 mmol) in THF (15 mL). Then the reaction mixture was cooled to 0 °C with stirring. DIAD (0.7 mL, 3.6 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room température for ovemight. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 84-F. LCMS-ESI+ (m/z): [M+H]+ found: 371.
Step 6
To a solution of 84-F (0.55 g, 1.5 mmol) EtOH (20 mL) was added hydrazine monohydrate (0.3 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 84G. LCMS-ESr (m/z): [M+H]+found: 241.
Step 7
A 100-mL round bottom flask was charged with 84-G (0.35 g, 1.4 mmol) and 84-H (0.5g, 1.4 mmol) in éthanol (10 mL). Sodium bicarbonate (0.24 g, 2.8 mmol) in water (10 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for ovemight. The mixture was diluted with EtOAc (50 mL) and washed with water (2 x). The aqueous fractions were extracted with EtOAc, and the organic fractions were combined, dried (NaîSOA, and concentrated. The crude 84-1 was used for next step without further purification. LCMS-ESI* (m/z): [M+H]+found: 583.
Step 8
A 100-mL rbf was charged with 84-1 (0.84 g, 1.4 mmol) in 4 N HCl /dioxane (8.2 mL). Then the reaction mixture was stirred at room température for 1 hour. After concentration, 0.74 g intermediate was obtained. The intermediate and DBU (1.1 g, 7.2 mmol) were dissolved in toluene (10 mL). The reaction mixture was heated to 110 °C with stirring for 1 hour. After concentration, the residue was purified by flash chromatography using hexanes - EtOAc as eluents to afford 84-J. LCMS-ESI* (m/z):
167 [M+H]+found: 409.
Step 9
A 100-mL round bottom flask was charged with 84-J (0.4 g, 0.98 mmol) in THF (5 mL) and MeOH (5 mL). 1 N KOH (3.0 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for 1 hour. The reaction mixture was acidified by adding 1 N HCl (3.0 mL). After concentration, the residue was co-evaporated with toluene (3 x). The crude acid, 2,4,6-trifluobenzylamine (0.32 g, 1.96 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.63 g, 4.9 mmol) and HATU (0.74 g, 1.9 mmol) were dissolved in DCM (10 mL). The reaction mixture was stirred at room température for 2 hours. The mixture was diluted with EtOAc (100 mL) and washed with saturated NaHCÛ3 (2x), saturated NH4CI (2x) and dried over Na2SÛ4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 84-K LCMS-ESI+ (m/z): [M+H]+found: 538.
Step 10
A 50-mL round bottom flask was charged with 84-K (0.5 g, 0.93 mmol) in TFA (6 mL). The reaction mixture was stirred at room température for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 84. 1H-NMR (400 MHz, Chloroform-d) δ 10.37 (s, IH), 8.28 (s, IH), 6.65 (t, J = 8.1 Hz, 2H), 4.80 (s, IH), 4.77 - 4.52 (m, 3H), 4.08 (d, J = 13.1Hz, IH), 3.88 (d, J =12.3 Hz, IH), 2.47 (d, J = 3.2 Hz, IH), 2.35 (s, IH), 2.16 (ddd, J = 14.3, 11.2, 3.6 Hz, IH), 1.93 - 1.57 (m, 3H), 1.29 - 1.19 (m, IH), 1.17 (d, J = 7.0 Hz, 3H). PF-NMR (376 MHz, Chloroform-d) δ -109.24 , -111.98. LCMS-ΕδΓ (m/z): found: 448.
Example 85
Préparation of Compound 85 (6aS,7R, 11 S)-1 -hydroxy-2,13-dioxo-N-(2,4,6-trifluorobenzyl)-6,6a,7,8,9,10,11,13octahydro-2H-7,ll-methanopyrido[r,2':4,5]pyrazino[l,2-a]azepine-3-carboxamide
168
1) H?, Pd/C, HCl
2) BOC2O
CICSOPh pyridine
Bu3SnH AIBN
COOEt
LiBH4
85-F
PhthNH
PPh3, DIAD
1) h2nnh2
COOEt
2) EK\ΛγΑ,θ , NaHCO3
O OBn 854________
3) HCl
4) DBU
Step 1
A solution of 85-A (1100 mg, 3.855 mmol) in DMSO (6 mL) and water (0.75 mL) was stirred at room température as N-iodosuccinmide (885 mg, 3.934 mmol) was added. After 2 h, additional N-iodosuccinmide (88 mg, 0.391 mmol) was added and the resulting mixture was stirred at room température for 1.5 h. The dark brown reaction 10 mixture was diluted with EtOAc, and washed with a mixture of 10 % aq. Na2S2O3
169 solution and aq. NaHCCh solution (~1:4 mixture) and then with water (with some brine). After the aqueous fractions were extracted with EtOAc, the organic fractions were combined, dried (Na2SO4), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to obtain 85-B. ’H-NMR (400 MHz, CDCh) δ 7.51 - 7.44 (m, 2H), 7.33 - 7.17 (m, 3H), 4.22 - 4.05 (m, 2H), 4.02 - 3.86 (m, 2H), 3.77 (d, J = 5.3 Hz, IH), 3.54 - 3.44 (m, IH), 3.27 (t, J = 4.5 Hz, IH), 2.75 - 2.66 (m, IH), 2.30 (dddd, J = 14.8, 13.1, 7.2, 5.8 Hz, IH), 2.14 (dddd, J = 14.8, 13.0,6.1,2.1 Hz, IH), 1.97 (d, J = 8.9 Hz, IH), 1.58 - 1.46 (m, IH), 1.45 - 1.34 (m, 4H), 1.24 (t, J = 7.1 Hz, 3H). LCMS-ESU (m/z): [M+H]+ calculated for C18H25INO3: 430.1; found: 430.0.
Step 2
A solution of 85-B (993 mg, 2.313 mmol), ΑΓΒΝ (305 mg, 1.857 mmol), and tributyltin hydride (1392 mg, 4.799 mmol) in toluene (15 mL) was stirred at 100 °C. After 2 h, the reaction mixture was cooled to room température, diluted with EtOAc, and washed with water and brine. After the aqueous fractions were extracted with EtOAc, the organic fractions were combined, dried (Na2SO4), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to obtain 85-C ^-NMR (400 MHz, CDCh) δ 7.57 - 7.49 (m, 2H), 7.32 - 7.23 (m, 2H), 7.23 7.15 (m, IH), 4.24-4.02 (m, 2H), 3.97 (q, J = 6.7 Hz, IH), 3.83 (d, J = 5.1 Hz, IH), 3.48 (t, J = 4.6 Hz, IH), 3.19 - 3.04 (m, IH), 2.58 (p, J = 4.0 Hz, IH), 2.30 (dddd, J = 14.7, 13.1, 7.0, 4.5 Hz, IH), 1.98 (d, J = 11.2 Hz, IH), 1.64 (tdd, J = 13.3, 6.2, 2.6 Hz, IH), 1.49 - 1.33 (m, 3H), 1.37 (d, J = 6.7 Hz, 3H), 1.32 - 1.26 (m, IH), 1.23 (t, J = 7.2 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+ calculated for C18H26NO3: 304.2; found: 304.1.
Step 3
A mixture of 85-C (725 mg, 2.39 mmol) and 20% Pd(OH)2/C (351 mg) in EtOH (25 mL) and 4 N HCl in dioxane (0.9 mL) was stirred under H2 atmosphère. After 2 h, the reaction mixture was filtered, and the filtrate was concentrated. LCMS-ESU (m/z): [M+H]+calculated for C10H18NO3: 200.13; found: 200.1. After the residue was co-evaporated with toluene (x 2), the residue and BOC2O (720 mg, 3.299 mmol) in THF (15 mL) was stirred at room température as Ν,Ν-diisopropylethylamine (DIPEA) (1.2 mL, 6.889 mmol) was added. After 1 h, the reaction mixture was diluted with water and
170 extracted with EtOAc (x 2). After the organic extracts were washed with water, the combined extracts were dried (Na2SO4> and concentrated. The residue was purified by flash using hexanes - EtOAc as eluents to obtain 85-D which appears to be a mixture of rotamers. Î-NMR (400 MHz, CDCh) δ 4.42 - 3.97 (m, 5H), 2.62 (d, J = 5.6 Hz, IH), 2.45 - 2.26 (m, IH), 2.25 - 2.15 (m, IH), 1.80 (td, J = 13.7, 6.7 Hz, IH), 1.66 (dd, J = 12.3, 6.6 Hz, 2H), 1.55 - 1.70 (m, 2H), 1.47 (s, 2H), 1.42 (s, 7H), 1.28 (dt, J = 9.5, 7.1 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+calculated for C15H26NO5: 300.2; found: 299.7.
Step 4
To a solution of 85-D (568 mg, 1.897 mmol) and pyridine (0.25 mL, 3.091 mmol) in THF (5 mL) was added phenyl chlorothionoformate (0.3 mL, 2.169 mmol) at 0 °C, which produced insoluble material quickly. After ~30 min at 0 °C, additional pyridine (0.3 mL, 3.709 mmol) and phenyl chlorothionoformate (0.3 mL, 2.169 mmol) were added. After 1.5 h at 0 °C and 1 h at room température, the mixture was concentrated, and the residue was dissolved in EtOAc and water. After séparation of two layers, the organic fraction was washed with ~0.1 N HCl, saturated aqueous NaHCÛ3, and brine . After the aqueous fractions were extracted with EtOAc, the combined organic fractions were dried (Na2SO4), and concentrated. The residue was purified by flash chromatography using EtOAc/hexanes as eluents to afford 85-E. ^-NMR (400 MHz, CDCh) δ 7.47 - 7.37 (m, 2H), 7.30 (t, J = 6.9 Hz, IH), 7.11 (dd, J = 8.0, 4.0 Hz, 2H), 5.54 (dt, J = 9.0, 4.9 Hz, IH), 4.50 (dt, J = 9.8, 5.3 Hz, IH), 4.35 (dd, J = 21.4, 5.0 Hz, IH), 4.30 - 4.14 (m, 2H), 2.71 (s, IH), 2.54 (s, IH), 2.14 - 2.00 (m, IH), 1.82 (m, 3H), 1.54 (m, IH), 1.48 (s, 4.5H), 1.45 (s, 4.5H), 1.30 (dt, J = 9.4, 7.1 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+calculated for C22H30NO6S: 436.2; found: 435.8.
Step 5
A mixture of 85-E (602 mg, 1.382 mmol), AIBN (182 mg, 1.108 mmol), and tributyltin hydride (608 mg, 2.096 mmol) in toluene (8 mL) was stirred at 100 °C. After 1 h, the reaction mixture was concentrated and the residue was dissolved in EtOAc before washing with water and brine. After the aqueous fractions were extracted with EtOAc, the combined organic fractions were dried (Na?SO4) and concentrated. The residue was purified with flash chromatography using EtOAc/hexanes as eluents to give
171
85-F which appears to be a mixture of rotamers. Î-NMR (400 MHz, CDCh) δ 4.37 4.06 (m, 4H), 2.69 - 2.53 (m, IH), 2.11 (m, IH), 1.97 (m,0.65H), 1.93 - 1.80 (m, 1.35H), 1.54 (s, 5H), 1.46 (s, 3.15H), 1.42 (s, 5.85H), 1.27 (m, 3H). LCMS-ESF (m/z): [MCÆL+HÈ calculated for ChHisNCU 228.1; found: 227.9.
Step 6
85-F (420 mg) was repurified and the purified 85-F in THF (3 mL) was stirred at 0 °C as 2.0 M LiBHj in THF (1.5 mL) was added. After 5 min, the mixture was stirred at room température for 17 h and additional 2.0 M L1BH4 in THF (1.5 mL) was added at room température. After 23 h at room température, additional 2.0 M L1BH4 in THF (3 mL) was added and the resulting mixture was stirred for ~72 h. After the reaction mixture was stirred at 0 °C as water was slowly added and further diluted with water, the product was extracted with EtOAc (x 2). The extracts were washed with water, combined, dried (Na2SÛ4), and concentrated. The residue was purified by flash chromatography using hexane - EtOAc as eluents to give 85-G. 1H-NMR (400 MHz, CDCh) δ 4.12 (t, J = 5.3 Hz, IH), 3.99 (dd, J = 12.0, 7.9 Hz, IH), 3.85 (dd, J = 8.0, 4.7 Hz, IH), 3.73 (dd, J = 11.9, 1.4 Hz, IH), 2.28 (d, J = 4.6 Hz, IH), 1.90 - 1.73 (m, 2H), 1.68 - 1.45 (m, 6H), 1.47 (s, 9H), 1.43 - 1.33 (m, IH). LCMS-ESF (m/z): [M-C4H8+H]+ calculated for C9H16NO3: 186.1; found: 186.0.
Step 7
A solution of 85-G (198 mg, 0.820 mmol), phthalimide (200 mg, 1.359 mmol), and PPI13 (488 mg, 1.861 mmol) in THF (10 mL) was stirred at 0 °C bath as DI AD (0.36 mL, 1.828 mmol) was added. After 30 min at 0 °C, the mixture was stirred at room température for 17 h. The reaction mixture was concentrated and the residue was purified by flash chromatography using hexane-EtOAc as eluents to 85-H which appears to be a mixture of rotamers. XH-NMR (400 MHz, CDCI3) δ 7.82 (dd, J = 5.4, 3.1 Hz, 2H), 7.69 (dd, J - 5.4, 3.1 Hz, 2H), 4.46 (s, IH), 4.19 (m, 2H), 3.95 (s, IH), 2.31 - 2.14 (m, IH), 2.05 (d, J = 16.5 Hz, IH), 1.84 (m, 2H), 1.79 - 1.70 (m, IH), 1.66 (m, IH), 1.61 - 1.30 (m, 12H). LCMS-ESF (m/z): [M+H]+calculated for C21H27N2O4: 371.2; found: 370.8.
Step 8
172
To a solution of 85-H (270 mg, 0.729 mmol) in EtOH (12 mL) was added hydrazine hydrate (0.145 mL, 3.083 mmol) atroom température and the resulting solution was stirred at 70 °C. After 1.5 h, the mixture was cooled to 0 °C and diluted with ether (30 mL) before stirring for 1 h at 0 °C. The mixture was filtered and the filtrate was concentrated. The residue was dissolved in CH2CI2 and filtered to remove some insoluble material. The resulting filtrate was concentrated. The residue, combined with 85-1 (257 mg, 0.742 mmol), and NaHCCh (131 mg, 1.559 mmol) in water (3 mL) and EtOH (3 mL) was stirred at room température. After 1 h, the mixture was diluted with water and extracted with EtOAc (x 2). After the extracts were washed with water, the organic extracts were combined, dried (Na2SO4), and concentrated. To a solution of the residue in CH2C12 (2 mL) was added 4 N HCl in dioxane (6 mL). After 1.5 h at room température, the solution was concentrated and co-evaporated with toluene. A mixture of the residue and DBU (0.6 mL, 4.012 mmol) in toluene (5 mL) was stirred at 100 °C bath. After 1 h, additional DBU (0.3 mL, 2.006 mmol) was added and the mixture was stirred another 1 h at 100 °C. After the mixture was concentrated, the residue was purified by flash chromatography using EtOAc - 20% MeOH/EtOAc as eluents to give 85-J. ^-NMR (400 MHz, CDCh) δ 8.08 (s, IH), 7.71 - 7.62 (m, 2H), 7.36 - 7.29 (m, 2H), 7.29 - 7.23 (m, IH), 5.44 (d, J = 9.8 Hz, IH), 5.10 (d, J = 9.8 Hz, IH), 4.44 - 4.28 (m, 3H), 4.23 (t, J = 13.0 Hz, IH), 3.99 (ddt, J = 10.2, 6.3, 3.6 Hz, 2H), 2.44 - 2.36 (m, IH), 2.29 (dt, J = 11.6, 5.3 Hz, IH), 1.84 (dt, J= 10.8, 5.3 Hz, 2H), 1.77 - 1.61 (m, 3H), 1.57 (d, J = 11.7 Hz, IH), 1.48 (ddd, J = 20.9, 12.3, 5.5 Hz, IH), 1.38 (t, J = 7.1 Hz, 3H). LCMS-ESI+ (m/z): [M +H]+calculated for C24H27N20s: 423.2; found: 423.3.
Step 9
A mixture of 85-J (214 mg, 0.507 mmol) in THF (4 mL) and MeOH (4 mL) was stirred at room température as 1 N KOH (1.1 mL) was added. After 30 min, the reaction mixture was concentrated to ~1 mL, acidified with 1 N HCl (~1.2 mL), and diluted with brine before extraction with CH2C12 (20 mL x 2). The combined extracts were dried (Na2SO4) and concentrated to obtain the crude acid. LCMS-ESC (m/z): [M +H]+calculated for C22H23N2O5: 395.2; found: 395.3.
A mixture of the crude acid (199 mg, 0.505 mmol), 2,4,6-trifluorobenzyl amine (130 mg, 0.807 mmol), and HATU (304 mg, 0.800 mmol) in CH2C12 (6 mL) was
173 stirred at room température as Ν,Ν-diisopropylethylamine (DIPEA) (0.62 mL, 3.559 mmol) was added. After 30 min, the reaction mixture was concentrated and the residue was dissolved in EtOAc, washed with saturated aqueous NH4CI (x 2), saturated aqueous NaHCÛ3 (x 2), and brine. After the aqueous fractions were extracted with EtOAc, two organic fractions were combined, dried (Na2SÜ4) and concentrated. The residue was purified by flash using EtOAc-20%MeOH/EA as eluents to obtain 85-K. ’H-NMR (400 MHz, CDCh) δ 10.40 (t, J = 5.7 Hz, IH), 8.42 (s, IH), 7.68 - 7.54 (m, 2H), 7.33 (ddd, J = 7.7, 6.3, 1.5 Hz, 2H), 7.30 - 7.26 (m, IH), 6.74 - 6.60 (m, 2H), 5.37 (d, J = 10.0 Hz, IH), 5.17 (d, J = 10.0 Hz, IH), 4.76 - 4.57 (m, 2H), 4.46 (dd, J = 6.0, 4.3 Hz, IH), 4.34 (t, J = 12.4 Hz, IH), 4.07 (dd, J = 12.4, 3.6 Hz, IH), 3.91 (dt, J = 12.4, 3.9 Hz, IH), 2.52 - 2.44 (m, IH), 2.32 (dd, J = 11.8, 6.2 Hz, IH), 1.92 (dt, J = 10.7, 5.4 Hz, IH), 1.83 - 1.70 (m, 3H), 1.67 (d, J = 11.7 Hz, IH), 1.52 (dddt, J = 25.5, 17.0, 11.8, 5.3 Hz, 2H). 19FNMR (376 MHz, CDCh) δ -109.15 (dq, J = 15.0, 7.5, 7.1 Hz, 1F), -111.85 (t, J = 6.8 Hz, 2F). LCMS-ESI+ (m/z): [M +H]+calculated for C29H27F3N3O4: 538.2; found: 538.3.
Step 10
85-K (187 mg, 0.348 mmol) was dissolved in trifluoroacetic acid (3 mL) at room température and stirred at room température. After 1 h, the solution was concentrated and the residue was dissolved in CH2CI2. After the solution was washed with 0.1 N HCl, the aqueous fraction was extracted with CH2CI2 (x 2). The organic fractions were combined, dried (Na2SÛ4), and concentrated. The residue was purified by flash chromatography using CH2Ch-20% MeOH in CH2CI2 as eluents to obtain 150 mg (96%) of compound 85. Compound 85 was further purified by recrystallization from methanol (10 mL) to give compound 85. *H-NMR (400 MHz, CDCh) δ 12.09 (s, IH), 10.39 (t, J = 5.7 Hz, IH), 8.36 (s, IH), 6.74 - 6.48 (m, 2H), 4.64 (d, J = 5.7 Hz, 2H), 4.59 (dd, J = 6.1, 4.4 Hz, IH), 4.36 - 4.18 (m, 2H), 4.12 (dt, J = 12.4, 4.1 Hz, IH), 2.68 - 2.47 (m, IH), 2.25 - 2.10 (m, IH), 2.10 - 1.98 (m, IH), 1.98 - 1.66 (m, 4H), 1.66 - 1.48 (m, 2H). 19F-NMR (376 MHz, CDCh) δ -109.23 (ddd, J = 15.1, 8.6, 6.0 Hz, 1F), -112.02 (t, J = 6.9 Hz, 2F). LCMS-ESI+ (m/z): [M +H]+ calculated for C22H21F3N3O4: 448.2; found: 448.3.
174
Example 86
Préparation of Compound 86 (lR,3S,4R,12aS)-7-hydroxy-3-methyl-6,8-dioxo-N-(2,4,6-trifluorobenzyl)1,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9carboxamide
COOMe
Bu3SnH
AIBN
COOMe
COOMe
Ph
86-C
1) H?, Pd/C, HCl
2) Boc2O
86-D
Dess-Ma rtin Periodinane
COOMe
Tebbe's reagent
86-E
COOMe
COOMe
86-G
86-H
PhthNH
PPh3, DIAD
1) H2NNH2 xCOOEt
0%
2)EtO. ,NaHCO3
O OBn 85-1
3) HCl
4) DBU
86-J
1) KOH _______________
2) 2,4,6-F3BnNH2, HATU, DIEA
86-K
Step 1
175
A solution of 86-A (10.160 g, 39.48 mmol) in DMSO (52 mL) and water (6.5 mL) was stirred at room température as N-iodosuccinmide (8.888 g, 39.50 mmol) was added. After 30 min, the dark brown reaction mixture was diluted with EtOAc, and washed with saturated aqueous NaHCO3 solution, 10 % aqueous Na2S2O3 solution ],and brine. After the aqueous fractions were extracted with EtOAc, the organic fractions were combined, dried (Na2SO4), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to obtain 86-B as a white solid. 1HNMR(400 MHz, CDC13) δ 7.33 - 7.19 (m, 5H), 4.25 - 4.12 (m, IH), 3.79 (q, J= 1.6 Hz, IH), 3.72 (q, J = 6.5 Hz, IH), 3.51 (s, IH), 3.47 (s, 3H), 3.31 (dd, J = 3.9, 1.6 Hz, IH), 2.76 - 2.69 (m, IH), 2.13 (ddd, J = 14.3, 7.8, 1.7 Hz, IH), 2.08 - 1.97 (m, IH), 1.91 (dtd, J = 14.1,4.0, 1.5 Hz, IH), 1.42 (d, J = 6.5 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+ calculated for Ci6H2iINO3: 402.1; found: 402.0.
Step 2
A solution of 86-B (12.468 g, 31.07 mmol), azobisisobutyronitrile (AIBN) (4.082 g, 24.86 mmol), and tributyltin hydride (18.047 g, 62.22 mmol) in toluene (150 mL) was stirred at 100 °C. After 30 min, the reaction mixture was cooled to room température, diluted with EtOAc, and washed with water and brine. After the aqueous fractions were extracted with EtOAc, the organic fractions were combined, dried (Na2SO4), and concentrated. The residue was purified by flash chromatography twice using hexanes - EtOAc as eluents to obtain 86-C. ’H-NMR (400 MHz, CDCI3) δ 7.39 7.31 (m, 2H), 7.31 - 7.24 (m, 2H), 7.24 - 7.17 (m, IH), 4.11 (s, IH), 3.72 (s, IH), 3.49 (s, 3H), 3.33 (d, J = 3.4 Hz, IH), 3.27 (d, J = 6.4 Hz, IH), 2.65 - 2.51 (m, IH), 1.92 (ddd, J = 13.6, 6.8, 2.4 Hz, IH), 1.69 - 1.50 (m, 2H), 1.47 (d, J = 10.1 Hz, IH), 1.41 (d, J = 6.6 Hz, 3H), 1.21 - 1.07 (m, IH). LCMS-ESI+ (m/z): [M+H]+calculated for Ci6H22NO3: 276.2; found: 276.1.
Step 3
A mixture of 86-C (4.187 g, 15.21 mmol) and 20% Pd(OH)2/C (1.022 g) in EtOH (100 mL) and 4 N HCl in dioxane (5.7 mL) was stirred under H2 atmosphère. After 1.5 h, the reaction mixture was filtered, and the filtrate was concentrated. After the residue was co evaporated with toluene, the residue was used
176 for the next step. LCMS-ESF (m/z): [M+H]+calculated for CsHuNCh: 172.1; found:
172.1.
After the residue was co-evaporated with toluene, the residue and BocîO (5.712 g, 26.17 mmol) in THF (45 mL) was stirred at room température as N,Ndiisopropylethylamine (DIPEA) (8 mL, 45.93 mmol) was added. After 30 min, the reaction mixture was diluted with water and extracted with EtOAc (x 2). After the organic extracts were washed with water, the combined extracts were dried (NazSCL) and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to obtain 86-D. H NMR spectrum suggests a mixture of rotamers. 1H-NMR (400 MHz, CDCb) Ô 4.20 (d, J = 7.6 Hz, IH), 4.19 - 4.10 (m, 2H), 4.08 (d, J = 3.5 Hz, IH), 3.72 (s, 3H), 2.74 (d, J = 5.6 Hz, IH), 1.97 (ddd, J = 13.6, 6.9, 2.8 Hz, IH), 1.88 1.78 (m, IH), 1.79 - 1.50 (m, IH), 1.46 (s, 3H), 1.38 (s, 6H), 1.31 (d, J= 13.3 Hz, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for Ci3H22NO5: 272.2; found: 271.6.
Step 4
A solution of 86-D (1659 mg, 6.115 mmol) in CH2CI2 (35 mL) was stirred at 0 °C bath as Dess-Martin periodinane (5.183 g, 12.22 mmol) was added in portions. After 5 min, the mixture was stirred at room température. After 2 h, the reaction mixture was cooled in an ice bath, quenched with water, and filtered. The filtrate was washed with saturated NaHCCh, dried (Na2SO4), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to give 86-E. ’H NMR spectrum suggests two rotamers. 1H-NMR (400 MHz, CDCb) δ 4.43 (d, J = 3.8 Hz, 0.5H), 4.39 (s, IH), 4.26 (s, 0.5H), 3.75 (s, 3H), 3.10 (s, IH), 2.24 (d, J = 4.5 Hz, 0.5H), 2.19 (d, J = 4.4 Hz, 0.5H), 2.12 (d, J == 4.4 Hz, 0.5H), 2.07 (d, J = 4.2 Hz, 0.5H), 2.01 (dd, J = 4.5, 2.2 Hz, 0.5H), 1.98 (dt, J = 4.3, 1.9 Hz, 0.5H), 1.80 (s, 0.5H), 1.77 (s, 0.5H), 1.46 (s, 4.5H), 1.40 (d, J - 2.8 Hz, 4.5H). LCMS-ESF (m/z): [M-C4H8+H]+ calculated for C9H12NO5: 214.1; found: 213.8.
Step 5
A solution of 86-E (528 mg, 1.961mmol) in THF (12 mL) was stirred at 0 °C as 0.5 M solution of Tebbe reagent in toluene (7.9 mL, 3.95 mmol) was added dropwise. After addition, the brown solution was allowed to warm to room température 177 slowly and was stirred at room température for 2.5 h. The reaction mixture was stirred at 0 °C bath as the reaction was quenched carefully by the addition of saturated aqueous NaHCCh solution. After the mixture was diluted with CH2CI2 and stirred at room température for 15 minutes, the resulting mixture was filtered through celite pad and the filter cake was washed with CH2CI2. After the two fractions in the filtrate were separated, the aq. fraction was extracted with CH2CI2, and the organic fractions were combined, dried (TfeSCU), and concentrated. The residue was purified by flash chromatography using hexanes - EtOAc as eluents to give 86-F. 'H NMR spectrum suggests two rotamers. 'H-NMR (400 MHz, CDCh) δ 5.13 (s, 0.6H), 5.04 (s, 0.4H), 4.82 - 4.71 (m, IH), 4.55 (s, 0.6H), 4.43 (s, 0.4H), 4.29 (d, J = 3.7 Hz, 0.4H), 4.24 (d, J = 3.7 Hz, 0.6H), 3.71 (s, 3H), 2.84 (s, IH), 2.14 (m, 2H), 1.75 (s, 0.6H), 1.74 - 1.70 (s, 0.4H), 1.55 (m, IH), 1.45 (s, 3.6H), 1.37 (s, 5.4H). LCMS-ESI+ (m/z): [M +H]+calculated for C14H22NO4: 268.2; found: 267.6.
Step 6
A mixture of 86-F (333 mg, 1.246 mmol) and 20% Pd(OH)2/C (53 mg) in EtOH (5 mL) was stirred under H2 atmosphère. After 30 min, the mixture was filtered and the filtrate was concentrated to give 86-G. 'H NMR spectrum suggests two rotamers. 'H-NMR (400 MHz, CDCh) δ 4.20 (m, IH), 4.08 (m, IH), 3.71 (two s, 3H), 2.68 (m, IH), 2.06 (m, IH), 1.80 - 1.63 (m, 2H), 1.63 - 1.51 (m, IH), 1.44 (s, 4H), 1.38 (s, 5H), 1.13 (m, 3H), 0.92 (m, IH). LCMS-ESI+ (m/z): [M +H]+ calculated for C14H24NO4: 270.2; found: 269.7.
Step 7
A solution of 86-G (336 mg, 1.482 mmol) in THF (5 mL) was stirred at 0 °C as 2.0 M L1BH4 in THF (1.5 mL) was added. After 5 min, the mixture was stirred at room température. After 2 h, additional 2.0 M LiBH4 in THF (1.5 mL) was added. After 21 h at room température, additional 2.0 M L1BH4 in THF (3 mL) was added. After 3 h at room température, the solution was heated at 35 °C for 18 h. The reaction mixture was cooled to 0 °C and quenched carefully with water. After the mixture was extracted with EtOAc (x 2), the two organic fractions were washed with water, combined, dried (Na2SO4), and concentrated. The residue was purified by flash chromatography using
178 hexanes - EtOAc to give 86-H ^-NMR (400 MHz, CDCh) δ 4.95 - 4.09 (br, IH), 4.05 (s, IH), 3.82 (dd, J = 11.5, 7.7 Hz, IH), 3.76 - 3.69 (m, IH), 3.66 (d, J = 11.5 Hz, IH), 2.45 (d, J = 4.1 Hz, IH), 2.03 (dqdd, J = 11.4, 7.0, 4.5, 2.6 Hz, IH), 1.77 - 1.57 (m, 2H), 1.48 (dd, J=10.1, 1.8 Hz, IH), 1.45 (s, 9H), 1.00 (d, J = 6.9 Hz, 3H), 0.93 (ddd, J =13.2,
4.7, 2.6 Hz, IH). LCMS-ESI+ (m/z): [M +H]+calculated for C13H24NO3: 242.2; found:
241.7.
Step 8
A solution of 86-H (218 mg, 0.903 mmol), phthalimide (218 mg, 1.482 mmol), and PPI13 (535 mg, 2.040 mmol) in THF (10 mL) was stirred at 0 °C bath as DIAD (0.40 mL, 2.032 mmol) was added. After 10 min at 0 °C, the mixture was stirred at room température for 19 h. The reaction mixture was concentrated and the residue was purified by flash chromatography using hexane-EtOAc as eluents to give 86-1. ’H NMR suggests two rotamers. ’H-NMR (400 MHz, CDCI3) δ 7.82 (dt, J = 7.3, 3.6 Hz, 2H), 7.70 (d, J = 5.3 Hz, 2H), 4.53 - 4.26 (m, IH), 4.26 - 3.89 (m, 2H), 3.89 - 3.65 (m, IH), 2.28 (m, IH), 2.04 (m, IH), 1.82 - 1.65 (m, 2H), 1.66 - 1.43 (m, 7H), 1.38 (s, 4H), 1.19 - 1.01 (m, 3H). LCMS-ESI+ (m/z): [M +H]+calculated for C21H27N2O4: 371.2; found: 370.8.
Step 9
To a solution of 86-1 (319 mg, 0.861 mmol) in EtOH (12 mL) was added hydrazine hydrate (0.17 mL, 3.494 mmol) at room température and the resulting solution was stirred at 70 °C bath. After 1.5 h, the mixture was cooled to 0 °C and diluted with ether (25 mL) before stirring for 1 h at 0 °C. The mixture was filtered and the filtrate was concentrated. The residue was dissolved in CH2CI2 and filtered to remove some insoluble material. The resulting filtrate was concentrated to give crude amine. LCMS-ESU (m/z): [M +H]+calculated for C13H25N2O2: 241.2; found: 240.9.
After the crude amine was co-evaporated with toluene, a mixture of the crude amine, 85-1 (300 mg, 0.866 mmol), and NaHCCh (150 mg, 1.845 mmol) in water (3 mL) and EtOH (3 mL) was stirred at room température. After 2 h, the mixture was diluted with water and extracted with EtOAc (x 2). After the extracts were washed with water, the organic extracts were combined, dried (Na2SO4), and concentrated. To a solution of the residue in CH2CI2 (2 mL) was added 4 N HCl in dioxane (6 mL). After
179
1.5 h at room température, the solution was concentrated and co-evaporated with toluene. A mixture of the residue and DBU (0.65 mL, 4.347 mmol) in toluene (6 mL) was stirred at 100 °C. After 1 h, additional DBU (0.65 mL, 4.347 mmol) was added and the mixture was stirred at 100 °C. Additional DBU (0.65 mL, 4.347 mmol) was added after 1 h and the mixture was stirred another 2.5 h at 100 °C. The mixture was diluted with CH2CI2 and washed with water containing 3 mL of 1 N HCl. The organic fraction was dried (Na2SO4) and concentrated. The residue was purified by flash chromatography using EtOAc-20% MeOH/EtOAc as eluents to give 86-J. 1H-NMR (400 MHz, CDCI3) δ 8.09 (s, IH), 7.70 - 7.62 (m, 2H), 7.37 - 7.27 (m, 3H), 5.48 (d, J = 9.9 Hz, IH), 5.16 (d, J = 9.9 Hz, IH), 4.53 (s, IH), 4.38 (m, 2H), 4.11 (m, IH), 3.97 (dd, J = 12.2, 3.0 Hz, IH), 3.88 (dt, J = 12.2, 3.0 Hz, IH), 2.63 (d, J = 4.2 Hz, IH), 2.28 (qd, J = 7.2, 3.1 Hz, IH), 2.00 1.88 (m, IH), 1.80- 1.56 (m,2H), 1.39 (t, J = 7.1 Hz, 3H), 1.07 (d, J = 6.9 Hz, 3H), 1.04 (dd, J = 5.0, 2.5 Hz, IH). LCMS-ESI+ (m/z): [M +H]+ calculated for C24H27N2O5: 423.2; found: 423.2.
Step 10
A mixture of 86-J (83 mg, 0.196 mmol) in THF (2 mL) and EtOH (2 mL) was stirred at room température as 1 N KOH (0.4 mL) was added. After 30 min, the reaction mixture was diluted with water and washed with CH2Q2. After the aqueous fraction was acidified with 1 N HCl 0.45 mL), the product was extracted with CH2C12 (x 2). The combined extracts were dried (Na2SO4) and concentrated to obtain the crude acid. LCMS-ESI+ (m/z): [M +H]+calculated for C22H23N2O5: 395.2; found: 395.2.
A mixture of the crude acid (69 mg, 0.175 mmol), 2,4,6-trifluorobenzyl amine (42 mg, 0.261 mmol), and HATU (106 mg, 0.279 mmol) in CH2C12 (3 mL) was stirred at room température as Ν,Ν-diisopropylethylamine (DIPEA) (0.25 mL, 1.435 mmol) was added. After 30 min, the reaction mixture was concentrated and the residue was dissolved in EtOAc, washed with saturated aqueous NH4C1 (x 2), saturated aqueous NaHCÛ3 (x 2), and brine. After the aqueous fractions were extracted with EtOAc, two organic fractions were combined, dried (Na2SO4) and concentrated. The residue was purified by flash chromatography using EtOAc-20%MeOH/EtOAc as eluents to obtain 86-K 1H-NMR (400 MHz, CDCh) δ 10.40 (t, J = 5.7 Hz, IH), 8.40 (s, IH), 7.66 - 7.51 (m, 2H), 7.36 - 7.29 (m, 2H), 7.29 - 7.23 (m, IH), 6.71 - 6.61 (m, 2H), 5.36 (d, J =
180
10.0 Hz, lH),5.18(d, J = 10.0Hz, IH), 4.73 - 4.58 (m, 2H), 4.53 (s, IH), 4.22-4.11 (m, IH), 4.03 (dd, J = 12.4,3.1 Hz, IH), 3.81 (dt, J = 12.3, 3.1 Hz, IH), 2.68 - 2.59 (m, IH), 2.29 (dddd, J = 11.4, 7.1,4.7, 2.4 Hz, IH), 1.94 (ddd, J =13.5, 11.2, 4.6 Hz, IH), 1.881.67 (m, 2H), 1.06 (d, J = 7.0 Hz, 3H), 1.03-1.09 (m, IH). 19F-NMR (376 MHz, CDCh) δ -109.14 (ddd, J = 15.2, 8.7, 6.2 Hz, 1F), -111.86 (t, J = 7.0 Hz, 2F). LCMS-ESE (m/z): [M +H]+calculated for C29H27F3N3O4: 538.2; found: 538.1.
Step 11
86-K (61 mg, 0.113 mmol) was dissolved in trifluoroacetic acid (2 mL) and stirred at room température. After 1 h, the solution was concentrated and the residue was dissolved in CH2CI2. After the solution was washed with 0.1 N HCl, the aqueous fraction was extracted with CH2CI2 (x 2). The organic fractions were combined, dried (Na2SÛ4), and concentrated. The residue was purified by flash chromatography using CH2Ch-20% MeOH in CH2CI2 as eluents to obtain compound 86. 1H-NMR (400 MHz, CDCh) δ 12.02 (s, IH), 10.40 (t, J = 5.7 Hz, IH), 8.35 (s, IH), 6.63 (t, J = 8.1 Hz, 2H), 4.62 (d, J = 5.7 Hz, 2H), 4.59 (s, IH), 4.22 (dd, J = 12.2, 3.5 Hz, IH), 4.13 (t, J = 11.9 Hz, IH), 4.05 (dt, J = 12.0, 3.1 Hz, IH), 2.77 - 2.70 (m, IH), 2.31 m, IH), 2.09 - 1.93 (m, IH), 1.93 - 1.81 (m, 2H), 1.10 (ddd, J = 13.9, 5.0, 2.1 Hz, IH), 1.02 (d, J = 6.9 Hz, 3H). 19F-NMR (376 MHz, CDCh) δ -109.22 (ddd, J = 15.1, 8.7, 6.1 Hz, 1F), -112.05 (t, J = 6.9 Hz, 2F). LCMS-ESI+ (m/z): [M +H]+ calculated for C22H21F3N3O4: 448.2; found: 448.3.
Example 87
Préparation of cA-5-aminotetrahydro-2H-pyran-3-ol
Step 1
A solution of benzyl (5-oxotetrahydro-2H-pyran-3-yl)carbamate (740 mg,
3.0 mmol) and cerium(III) chloride heptahydrate (1.12 g, 3.0 mmol) in 20 mL methanol
181 was cooled to 0 °C and sodium borohydride (120 mg, 3.2 mmol) was then added portionwise. The reaction mixture was allowed to stir at 0 °C for 45 minutes and then quenched by slow addition of 1 mL acetone followed by 3 hours stirring at room température. The reaction mixture was partitioned between water and dichloromethane and the aqueous phase extracted into dichloromethane followed by 2-butanol. The combined organic phases were dried over magnésium sulfate, filtered, concentrated, and the residue purified by flash chromatography (0-100% EtOAc/hexanes) to afford the desired czx-benzyl ((3R,5S)-5-hydroxytetrahydro-2H-pyran-3-yl)carbamate. 1H-NMR (400 MHz, Chloroform-cQ δ 7.39 - 7.26 (m, 5H), 6.06 (br s, IH), 5.07 (s, 2H), 3.86 3.70 (m, 2H), 3.69 - 3.47 (m, 4H), 2.00 - 1.89 (m, IH), 1.76 (d, J= 13.5 Hz, IH). The undesired //zzzz.s-isomer was also isolated.
Step 2
To a solution of m-benzyl ((3R,5S)-5-hydroxytetrahydro-2H-pyran-3yl)carbamate (290 mg, 1.16mmol)in5 mL 1:1 DCM:EtOH was added 10wt%Pd/C (255 mg). This mixture was stirred under balloon pressure hydrogen for 18 hours and palladium removed by filtration thru celite with éthanol rinse. Upon concentration of filtrate, the czs-5-aminotetrahydro-2H-pyran-3-ol was afforded and carried on as crude.
Example 88
Préparation of Compound 88 '(2R, 5 S, 13 aR)-N-(3 -chloro-2-fluorobenzyI)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13 aoctahydro-2,5-methanopyrido[ 1 ',2' :4,5]pyrazino[2,1 -b] [ 1,3]oxazepine- 10-carboxamide
Compound 88 was prepared in a similar manner to compound 15 using (3chloro-2-fluorophenyl)methanamine in place of (4-fluorophenyl)methanamine. Ή
182
NMR (400 MHz, Chloroform-7) δ 10.43 (br s, IH), 8.34 (br s, IH), 7.32 - 7.24 (m, 2H), 7.02 (t, J= 7.9 Hz, IH), 5.36 (d, J= 9.4 Hz, IH), 5.30 (s, 2H), 4.70 (d, J= 6.0 Hz, 3H), 4.24 (d, J= 12.0 Hz, IH), 4.00 (dd, 7 = 12.7, 9.5 Hz, IH), 2.18 - 1.96 (m, 4H), 1.96 1.83 (m, IH), 1.60 (dt, 7= 12.4, 3.1 Hz, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for
C21H19CIFN3O5: 448.11; found: 448.2.
Example 89
Préparation of Compound 89 (2R, 5 S, 13 aR)-N-(2,5-difluorobenzyl)-8-hydroxy-7,9-dioxo-2,3,4,5,7,9,13,13 aoctahydro-2,5-methanopyrido[l',2':4,5]pyrazino[2,l-b][l,3]oxazepine-10-carboxamide
Compound 89 was prepared in a similar manner to compound 15 using (2,5-difluorophenyl)methanamine in place of (4-fluorophenyl)methanamine. 1H-NMR (400 MHz, Chloroform-ti) δ 10.32 (t, 7= 5.8 Hz, IH), 8.31 (br s, IH), 7.15 - 6.89 (m, 2H), 6.86 (d, 7= 8.5 Hz, IH), 5.40 (d, 7= 9.3 Hz, IH), 5.24 (s, IH), 4.67 - 4.51 (m, 3H), 4.35 - 4.28 (m, IH), 3.99 - 3.90 (m, IH), 2.16 - 1.85 (m, 5H), 1.60 - 1.50 (m, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H19F2N3O5: 432.14; found: 432.2.
Example 90
Préparation of Compound 90 (lR,4S,12aR)-N-(3-chloro-2-fluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: r,2'-d]pyrazine-9-carboxamide
183
Compound 90 was prepared in a similar manner to compound 41 using (3chloro-2-fluorophenyl)methanamine in place of (2,4,6-trifluorophenyl)methanamine. ’H-NMR (400 MHz, Chloroform-tf) 5 9.22 (s, IH), 8.79 (s, IH), 7.39 - 7.28 (m, 2H), 7.06 (t, 8.0 Hz, IH), 4.89 (s, IH), 4.70 - 4.56 (m, 3H), 4.06 - 3.83 (m, 2H), 3.04 2.88 (m, IH), 2.77 (s, IH), 1.97 - 1.58 (m, 6H). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H19CIFN3O4: 432.11; found: 432.2.
Example 91
Préparation of Compound 91 (1R,4S, 12aR)-7-hydroxy-6,8-dioxo-N-(2,3,4-trifluorobenzyl)-l,2,3,4,6,8,12,12aoctahydro-l,4-methanodipyrido[l,2-a:l',2'-d]pyrazine-9-carboxamide
Compound 91 was prepared in a similar manner to compound 41 using (2,3,4-trifluorophenyl)methanamine in place of (2,4,6-trifluorophenyl)methanamine. *HNMR (400 MHz, Chloroform-tT) δ 10.25 (s, IH), 8.45 (s, IH), 7.10 (d, J= 5.1 Hz, IH), 6.90 (d, J= 8.7 Hz, IH), 4.89 (s, IH), 4.63 (s, 2H), 4.22 (d, J= 11.6 Hz, IH), 3.93 - 3.73 (m, 2H), 2.71 (s, IH), 1.97 - 1.57 (m, 6H). LCMS-ESH (m/z): [M+H]+ calculated for C21H18F3N3O4: 434.13; found: 434.2.
184
Example 92
Préparation of Compound 92 (1R,4S, l2aR)-N-(4-chloro-2-fluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
Compound 92 was prepared in a similar manner to compound 41 using (4chloro-2-fluorophenyl)methanamine in place of (2,4,6-trifluorophenyl)methanamine. ’H-NMR (400 MHz, Chloroform-rf) δ 10.28 (s, IH), 8.41 (s, IH), 7.29 (s, IH), 7.11 6.95 (m, 2H), 4.85 (s, IH), 4.57 (s, 2H), 4.22 (d, J= 10.2 Hz, IH), 3.81 (q, J= 13.9, 13.1 Hz, 2H), 2.68 (s, IH), 1.99 - 1.50 (m, 6H). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H19CIFN3O4: 432.11; found: 432.2.
Example 93
Préparation of Compound 93 (1R,4S, 12aR)-N-(2-chloro-4,6-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-methanodipyrido[ 1,2-a: 1 ’,2'-d]pyrazine-9-carboxamide
185
Step 1
A 5 mL microwave vial was charged with 2-bromo-l-chloro-3,5difluorobenzene (540 mg, 2.4 mmol), cuprous cyanide (436 mg, 4.87 mmol), tetrakis(triphenylphosphine)palladium (63 mg, 0.05 mmol), sealed, and evacuated/backfilled with nitrogen. To this was added 5 mL degassed DMF. The sealed vessel was heated at 110 °C for 18 hours, diluted with ethyl acetate, and washed sequentially with twice 9:1 NH4OH:NH4Cl(aq), twice 5% LiCl(aq), and brine. The organic phase was then dried over magnésium sulfate, filtered, and concentrated. The crude residue was purified by flash chromatography (100% hexanes) to afford 2-chloro-4,6difluorobenzonitrile. flH-NMR (400 MHz, Chloroform-rf) δ 7.13 (dt, J= 8.0, 1.9Hz, IH), 6.93 (td, 8.5, 2.3 Hz, IH).
Step 2
To a solution of 2-chloro-4,6-difluorobenzonitrile (210 mg, 1.2 mmol) in 2.4 mL THF was added a 2M solution of borane-DMS in THF (0.6 mL). This reaction mixture was allowed to stir at refluxing température for 18 hours resulting in a loss of ail solvent. The residue was re-dissolved in 3 mL THF, cooled to 0 °C, a 6M solution of HCl(aq) was carefully added, and the mixture returned to reflux for 30 minutes. The
186 reaction mixture was once again cooled to 0 °C and treated with 4M NaOH(aq). The aqueous phase was extracted with DCM, combined organic phases dried over magnésium sulfate, filtered, and concentrated. The crude residue was purified by flash chromatography (0-10% MeOH/DCM) to afford (2-chloro-4,6difluorophenyl)methanamine. ’H-NMR (400 MHz, Chloroform-7) δ 6.95 (dt, J= 8.3, 2.1 Hz, IH), 6.76 (td, J= 9.4, 2.5 Hz, IH), 3.94 (d, J= 1.9 Hz, 2H).
Steps 3 and 4
A solution of 93-A (74 mg, 0.11 mmol), (2-chloro-4,6difluorophenyl)methanamine (48.5 mg, 0.27 mmol), HATU (100 mg, 0.26 mmol), and
N, N-diisopropylethylamine (0.1 mL, 0.57 mmol) in 1 mL dichloromethane was stirred at room température for one hour at which point complété disappearance of 93-A and formation of 93-B was observed by LCMS. TFA (0.65 M) was added and the mixture was stirred at room température for one hour, at which point 1 mL DMF was added. The reaction mixture and then concentrated and purified by préparative HPLC (ACN/H2O +
O. 1% TFA) to afford compound 93. ’H-NMR (400 MHz, DMSO-îA) δ 10.41 (t, J = 5.Ί Hz, IH), 8.33 (s, IH), 7.41 - 7.26 (m, 2H), 4.72 - 4.57 (m, 3H), 4.43 (dd, 7= 12.5, 3.6 Hz, IH), 3.94 (t, 7= 12.4 Hz, 2H), 3.77 (dd, 7= 12.4, 3.6 Hz, 3H), 1.87 - 1.67 (m, 3H), 1.67 - 1.45 (m, 2H), 1.43 (d, 7= 10.4 Hz, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H18CIF2N3O4·. 450.10; found: 450.2.
Example 94
Préparation of Compound 94 (lR,4S,12aR)-N-benzyl-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12a-octahydro-l,4methanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
187
Compound 94 was prepared in a similar manner to compound 41 using phenylmethanamine in place of (2,4,6-trifluorophenyl)methanamine. ’H-NMR (400 MHz, Chloroform-d) 5 10.37 (s, IH), 8.26 (s, IH), 7.37 - 7.19 (m, 5H), 4.55 (d, J= 4.8 Hz, IH), 4.34 (d, J= 5.7 Hz, IH), 4.23 (d, J= 9.8 Hz, IH), 4.09 (d, J= 28.2 Hz, IH), 3.78 (d,J= 10.9 Hz, IH), 3.64 (d, J= 13.2 Hz, IH), 3.14 - 3.01 (m, IH), 1.91-1.49 (m, 4H). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H21N3O4: 380.16; found: 380.2.
Example 95
Préparation of chiral tert-butyl 3-((1,3-dioxoisoindolin-2-yl)methyl)-2azabicyclo[2.1.1]hexane-2-carboxylates 95-A and 95-B
Chiral HPLC
Lux Cellulose-2
Absolute stereochemistries unknown
Step 1
To a 0 °C solution of racemic tert-butyl 3-(hydroxymethyl)-2azabicyclo[2.1.1]hexane-2-carboxylate (285 mg, 1.34 mmol), triphenylphosphine (425 mg, 1.62 mmol), and phthalimide (240 mg, 1.62 mmol) in 9 mL THF was added dropwise a solution of diisopropyl azodicarboxylate (0.35 mL, 1.8 mmol) in 1 ml THF. The reaction mixture was warmed to room température, stirred for 90 minutes, concentrated onto silica, and purified by flash chromatography (0-25% EtOAc/hexanes) to afford tertbutyl 3-((1,3-dioxoisoindolin-2-yl)methyl)-2-azabicyclo[2.1. l]hexane-2-carboxylate as a racemic mixture. LCMS-ESI+ (m/z): [M+H]+ calculated for C19H23N2O4: 343.2; found: 342.8.
Step 2
188
Racemic tert-butyl
-(( 1,3 -dioxoisoindolin-2-yl)methy 1)-2azabicyclo[2.1.1]hexane-2-carboxylate (655 mg, 1.91 mmol) was separated by chiral HPLC on a Lux Cellulose-2 column using an acetronitrile eluent to afford chiral 95-A (first eluting peak) and 95-B (second eluting peak) in enantioenriched form. For 95-A: 5 144 mg, 98%ee (absolute stereochemistry unknown). For 95-B: 242 mg, 49%ee (absolute stereochemistry unknown).
Example 96
Préparation of Compound 96 (1R,3R, 1 laS)-6-hydroxy-5,7-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,5,7,11,11ahexahydro-1 H-1,3 -methanopyrido [ 1,2-a]pyrrolo[ 1,2-d]pyrazine-8-carboxamide
O OH
189
1) KOH
2) HATU, DIEA
2,4,6-F3BnBr
(Absolute stereochemistries unknown)
Step 1
To a solution of intermediate 95-A (141 mg, 0.41 mmol, 98% ee, unknown absolute stereochemistry) in 9 mL éthanol was added hydrazine hydrate (0.5 mL, 10.3 mmol) and stirred at 70 °C for 18 hours to afford 96-A of unknown absolute stereochemistry. Solids were removed by filtration and the filtrate concentrated and carried on as crude.
Step 2
A mixture of crude 96-A (0.41 mmol assumed), 96-B (430 mg, 1.25 mmol), and sodium bicarbonate (69 mg, 0.82 mmol) in 2 mL water and 2 mL éthanol were stirred at room température for 18 hours, after which the reaction mixture was diluted with water and thrice extracted to ethyl acetate. The combined organic phases 15 were dried over magnésium sulfate, filtered, concentrated. The crude residue (222 mg) was dissolved in 1.5 mL DCM and 4 N HCl in dioxane (4 mL) was added and stirred for 90 minutes at room température. The mixture was concentrated to dryness and
190 coevaporated with toluene. The crude residue and DBU (0.3 mL, 2.0 mmol) in 6 mL methanoi was stirred at 50 °C for 90 minutes. The reaction mixture was then concentrated onto silica gel and purified by flash chromatography (0-10% MeOH/DCM) to afford 96C LCMS-ESI+ (m/z): [M+H]+ calculated for C22H22N2O5: 395.16; found: 395.2.
Step 3
A mixture of 96-C (112 mg, 0.28 mmol), IM aqueous potassium hydroxide (1 mL), 4 mL methanol, and 4 mL THF was stirred at room température for 3 hours, at which point the mixture was diluted with dichloromethane, acidifîed by addition of IM aqueous hydrogen chloride, and the organic phase extracted to dichloromethane. The combined organics were dried, filtered, and concentrated from toluene. After drying under vacuum, the residue was suspended in 1.5 mL DCM and trifluorobenzylamine (62 mg, 0.38 mmol), HATU (220 mg, 0.58 mmol), and Ν,Ν-diisopropylethylamine (DIPEA) (0.15 mL, 0.86 mmol) were added. This reaction mixture was stirred at room température for 2 hours to afford 96-D which was carried forward as crude.
Step 4
Trifluoroacetic acid (1.7 mL, 22.2 mmol) was added to the crude reaction solution containing 96-D from the prior step and the reaction mixture allowed to stir at room température for 90 minutes. 1 mL of DMF was then added, the reaction mixture concentrated down to ~1 mL, filtered, and purified by préparative HPLC (ACN/water + 0.1% TFA) to afford compound 96 (unknown absolute stereochemistry). Î-NMR (400 MHz, DMSOé) δ 10.45 - 10.35 (m, IH), 8.39 (s, IH), 7.23 - 7.09 (m, 2H), 4.67 (dd, J = 12.6, 4.8 Hz, 2H), 4.53 (d, J= 5.5 Hz, 2H), 4.20 (dd, J= 11.9, 3.8 Hz, IH), 4.05 - 3.95 (m, IH), 2.96 - 2.88 (m, IH), 2.16 (d, J= 7.0 Hz, IH), 1.97 (d, J= 7.0 Hz, IH), 1.68 1.60 (m, IH), 1.53 - 1.45 (m, IH). LCMS-ESU (m/z): [M+H]+ calculated for C20H16F3N3O4: 420.12; found: 420.2.
Example 97
Préparation of Compound 97 (1 S,3 S, 11 aR)-6-hydroxy-5,7-dioxo-N-(2,4,6-trifluorobenzyl)-2,3,5,7,11,11ahexahydro- IH-1,3 -methanopyrido[ 1,2-a]pyrrolo[ 1,2-d]pyrazine-8-carboxamide
191
O OH (Absolute stereochemistry unknown)
Compound 97 (49% ee, unknown absolute stereochemistry) was prepared in a similar manner to compound 96 using intermediate 95-B (49% ee, unknown absolute stereochemistry) in place of enantiomerically opposite intermediate 95-A. *H-NMR (400 MHz, DMSO-^e) δ 10.39 (t, J= 5.7 Hz, IH), 8.42 (s, IH), 7.25 - 7.13 (m, 2H), 4.73 4.66 (m, 2H), 4.54 (d, 5.7 Hz, 2H), 4.20 (dd, J= 12.3, 3.9 Hz, IH), 4.01 (t, J= 12.4
Hz, IH), 2.93 (dd, J= 6.7, 3.4 Hz, IH), 2.19 - 2.14 (m, IH), 1.97 (d, J= 8.3 Hz, IH), 1.65 (dd, .7 = 10.4, 7.9 Hz, IH), 1.49 (dd, J = 10.5, 7.7 Hz, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C2oHi6F3N304: 420.12; found: 420.2.
Example 98
Préparation of Compound 98 (lS,4R,12aR)-3,3-difluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)l,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a:r,2'-d]pyrazine-9carboxamide
192
4)
Mitsunobu
6) NaHCO3
7) 4N HCl in dioxane
8) DBU/MeOH
98-F
Step 1
98-A (0.5g, 1.87mmol) was dissolved in DCM (20 mL) and cooled to 0 °C under Nitrogen. Dess-Martin Periodinane (1.59 g, 3.74 mmol) was added slowly. The mixture was stirred for 2 h at room température, quenched with Na2S2O3/NaHCCb (7:1) aqueous saturated solution (160 mL) and stirred vigorously until two layers were separated. The crude product was twice extracted with DCM. The combined organic layers was dried over sodium sulfate and concentrated. The crude product was purified 10 by flash chromatography on silica gel with 0-20%MeOH/DCM to afford 98-B. ’H-NMR (400 MHz, Chloroform-d) δ 4.34 - 4.05 (m, IH), 3.97 - 3.75 (m, IH), 3.69 (s, 3H), 2.89 (dd, J = 4.4, 2.1 Hz, IH), 2.30 - 1.97 (m, 3H), 1.56 (d, J = 11.3 Hz, IH), 1.35 (s, 9H). LCMS-ESI+ (m/z)·. [M+H]+calculated for C13H19NO5: 269.13; found: 270.78.
Step 2
A solution of 98-B (504 mg, 1.87mmol) in DCM (15 mL) was stirred at 0 °C. DAST (1ml) was added drop wise to the reaction mixture. After ovemight stirring
193 at room température, the reaction mixture was cooled back to 0 °C. Saturated NaHCCh (10 mL) was added slowly. The mixture was extracted with twice with DCM and dried over Na2SC>4. After concentrating, the residue was purified by flash chromatography 050% EtOAc/hexanes to give 98-C. 1H-NMR (400 MHz, Chloroform-d) δ 4.45 - 4.18 (m, IH), 3.85 (m, IH), 3.72 (d, J = 1.5 Hz, 3H), 2.72 (ddd, J = 5.1, 3.2, 1.6 Hz, IH), 2.27 1.52 (m, 4H), 1.41 (d, J = 21.9 Hz, 9H). 19F-NMR (376 MHz, Chloroform-d) δ -91.72 -93.99 (m), -113.65 - -115.98 (m). LCMS-ESI+ (m/z): [M+H]+ calculated for C13H19F2NO4: 291.13; found: 291.55.
Step 3
98-C (476 mg, 1.634mmol) in THF (20 mL) was stirred at 0 °C as 2.0 M L1BH4 in THF (2.4 mL, 4.8mmol) was added. The mixture was warmed to room température and stirred for 4 h. The reaction mixture was quenched with ice and diluted with EtOAc and saturated NH4CI (some H2 évolution). After the two phases were separated, the organic fraction was washed with brine, dried (Na2SO4), and concentrated. The crude product of 98-D was used as is for the next step. LCMS-ESI+ (m/z): [M+H]+calculated for C12H19F2NO3: 263.13; found: 164.10.
Step 4
98-D (1.634mmol), phthalimide (0.36 g, 2.4 5mmol), and PPI13 (0.855 g, 3.26mmol) in THF (10 mL) was stirred at 0 °C bath as DIAD (0.642 mL, 3.26mmol) was added. After addition, the mixture was stirred at 0 °C for 30 min and then at room température for 16 h. It was diluted with EtOAc, and saturated NH4C1. After stirring for 5 min, a solid was filtered off and the two phases were separated. The organic phase was washed with brine, dried (Na2SO4), and concentrated. The crude product was purified by flash chromatography with 0-50%EA/Hex as eluents to give 98-E. ’H-NMR suggests a mixture oftwo rotamers. 1H-NMR(400 MHz, Chloroform-d) δ 7.89 - 7.80 (m, 2H), 7.78 - 7.66 (m, 2H), 5.02 (ddt, J = 16.6, 12.5, 6.3 Hz, IH), 4.24 (d, J = 71.8 Hz, IH), 4.10 3.92 (m, IH), 3.83 - 3.51 (m, 2H), 2.46 (s, IH), 2.21 - 1.98 (m, 2H), 1.87 - 1.62 (m, 2H),1.31 (d, J = 8.5 Hz, 9H); 19F-NMR (376 MHz, Chloroform-d) δ -91.22 - -93.58 (m), -113.20 - -115.45 (m). LCMS-ESI+ (m/z): [M+H]+calculated for C20H22F2N2O4: 392.15; found: 393.3.
194
Step 5
To a solution of 98-E (696 mg, 1.774mmol) in EtOH (lOmL) was added hydrazine hydrate (lmL) at room température and the resulting solution was stirred at room température for 2 h. The mixture was diluted with ethyl ether (30 mL) and stirred at 0 °C for 60 min before filtration. The filtrate was concentrated and the residue was dissolved in CH2CI2 and filtered. The filtrate was concentrated and purified by flash chromatography on silica gel with 0-20% MeOH (0.2% TEA) /DCM to give 98-F. NMR (400 MHz, Chloroform-d) δ 4.91 (p, J = 6.2 Hz, IH), 4.29 - 3.97 (m, IH), 3.36 2.93 (m, 2H), 2.49 (qt, J - 8.8, 5.2 Hz, 2H), 2.08 (dddd, J = 25.5, 14.0, 7.1, 4.9 Hz, IH), 1.89 - 1.49 (m, 4H), 1.41 and 1.21 (d, J = 6.2 Hz, 9H). 19F-NMR (376 MHz, Chloroformd) δ -91.63 - -93.16 (m), -113.11 - -115.08 (m). LCMS-ESI+ (m/z): [M+H]+calculated for C12H20F2N2O2: 262.15; found: 262.8.
Step 6, 7 and 8
The mixture of 98-G (375.8 mg, 1.55 mmol), 98-E (370 mg, 1.41 mmol), andNaHCO3 (261 mg, 3.10 mmol) in water (5 mL) and EtOH (5 mL) was stirred at room température for 2 h. The mixture was diluted with brine and extracted with EtOAc (x 2). The extracts were combined, dried (Na2SO4), concentrated, and dried in vacuo to afford crude A. LCMS-ESI+ (m/z): [M+H]+ 591.59. Crude A (1.38mmol) in CH2CI2 (5 mL) was added 4 N HCl in dioxane (5 mL). After 2 h at room température, mixture was concentrated to dryness. It was co-evaporated with toluene once and dried in vacuo to afford crude B. B (1.38mmol + 0.442mmol) and DBU (3 mL, 1 Immol) in anhydrous MeOH ( 15 mL) were stirred at 50 °C bath for 40 min. The mixture was concentrated. The residue was purified by flash chromatography (80 g column) using 0 - 20% MeOH/DCM as eluents to give 98-H. LCMS-ESI+ (m/z): [M+H]+ calculated for C23H22F2N2O5: 444.15; found: 445.36 (90%), 431.18 (10%).
Steps 9.10 and 11
The remaining steps were performed using procedures similar to Example 41 to afford desired compound 98. *H-NMR (400 MHz, Chloroform-d) δ 10.29 (d, J = 6.1 Hz, IH), 8.34 (s, IH), 6.65 (dd, J = 8.7, 7.5 Hz, 2H), 4.83 (s, IH), 4.72 - 4.58 (m, 2H),
195
4.36 - 4.10 (m, 2H), 4.05 (t, J = 11.5 Hz, IH), 2.97 (d, J = 4.4 Hz, IH), 2.49 - 2.08 (m, 3H), 2.12-1.94 (m, IH). 19F-NMR (376 MHz, Chloroform-d) δ -92.08 - -93.57 (m, 1F), -108.92 (ddd, J = 15.1, 8.8, 6.3 Hz, 1F), -109.30 - -110.65 (m, 1F), -112.16 (p, J = 7.3 Hz, 2F). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H16F5N3O4: 469.11; found: 5 470.23.
Example 99
Préparation of Compound 99 (lR,3S,4R,12aR)-7-hydroxy-3-methyl-6,8-dioxo-N-(2,4,6-trifluorobenzyl)1,2,3,4,6,8,12,12a-octahydro-1,4-methanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-910 carboxamide
196 +
6) NaHCO3
7) HCl
8)DBU
9) LiOH
10) HATU
11) TFA
Step 1
To a stirred solution of 99-A (1 g, 3.71 mmol) in THF (20 mL) was added dropwise a solution of the Tebbe reagent (0.5 M in toluene, 14.85mL, 7.42 mmol) at 0 °C. After addition, the brown solution was allowed to warm to room température slowly and was stirred at room température for 2 h. The reaction was quenched carefully by the addition of saturated NaHCCL solution at 0 °C, and the mixture was stirred at room température for 15 minutes. The mixture was filtered through celite, and the filter cake was washed with ether and DCM (1:1) twice. After separated layers, the organics were combined and concentrated in vacuo, and the residue was purified by column chromatography on silica gel column with 0-50% EtOAc/hexanes to afford 99-B. 1HNMR (400 MHz, Chloroform-d) δ 5.06 (dt, J = 48.6, 2.6 Hz, IH), 4.73 (d, J = 7.0 Hz, IH), 4.42 (d, J = 61.8 Hz, IH), 3.81 (d, J = 48.2 Hz, IH), 3.73 (d, J = 1.6 Hz, 3H), 2.74 (dd, J = 9.4, 4.4 Hz, IH), 2.38 (ddt, J = 13.5, 4.5, 2.5 Hz, IH), 2.18 - 2.06 (m, IH), 1.99 (dt, J = 10.2, 2.4 Hz, IH), 1.58 (s, IH), 1.42 (d, J = 25.5 Hz, 9H). LCMS-ESU (m/z): [M+H]' calculated for C14H21NO4: 267.15; found: 267.65.
Step 2
A mixture of 99-B (675 mg, 2.506 mmol) and 20% Pd(OH)2/C (500 mg) in EtOH (50 mL) was stirred under H2 atmosphère. The mixture was filtered through Celite and the filtrate was concentrated to give 99-C. ’H-NMR (400 MHz, Chloroformd) δ 4.23 - 3.99 (m, IH), 3.77 - 3.64 (m, 4H), 2.55 (d, J = 4.8 Hz, IH), 2.14 - 1.86 (m, 3H), 1.42 (d, J = 24.2 Hz, 9H), 0.96 (d, J = 6.6 Hz, 3H), 0.85 (ddd, J = 12.5, 4.8, 2.4 Hz, IH). LCMS-EST+ (m/z): [M+H]+calculated for C14H23NO4: 269.16; found: 269.69.
197
Step 3
99-C (670 mg, 2.488 mmol) in THF (20 mL) was stirred at 0 °C as 2.0 M LiBH4 in THF (3.7mL, 7.46 mmol) was added. The mixture was warmed to room température and stirred for 4h. The reaction mixture was quenched with ice and diluted with EtOAc and saturated NH4CI (some H2 évolution). After two phases were separated, the organic fraction was washed with brine, dried (Na2SO4), and concentrated. The crude alcohol 99-D was used as is for the next step. LCMS-ESI+ (m/z): [M+H]+ calculated for C13H23NO3: 241.17; found: 241.76.
Steps 4 and 5
Steps 4 and 5 were performed using procedures similar to those in Example 41 to afford 99-F. LCMS-ESI+ (m/z): [M+H]+ calculated for C13H24N2O2: 240.18; found: 241.2.
Step 6, 7 and 8
Steps 6, 7 and 8 were performed using procedures similar to that of Example 41 to give 99-G. LCMS-ESI+ (m/z): [M+H]1 calculated for C24H26N2O5: 422.18; found: 423.21.
Step 9, 10 and 11
The remaining steps were performed using procedures similar to Example 41 to afford compound 99. 'H-NMR (400 MHz, Chloroform-d) 5 11.71 (s, IH), 10.36 (t, J = 5.7 Hz, IH), 8.28 (s, IH), 6.63 (t, J = 8.1 Hz, 2H), 4.63 (t, J = 5.4 Hz, 3H), 4.12 (dd, J = 12.3, 3.5 Hz, IH), 3.83 (t, J = 12.3 Hz, IH), 3.67 (dd, J = 12.3, 3.4 Hz, IH), 2.64 2.52 (m, IH), 2.30 (ddq, J= 10.5, 7.2, 3.6 Hz, IH), 2.13 (td, J= 12.1, 4.4 Hz, IH), 1.82 - 1.63 (m, 2H), 1.24 (d, J = 3.3 Hz, IH), 1.04 (d, J = 6.9 Hz, 4H), 0.90 - 0.79 (m, IH). 19F-NMR (376 MHz, Chloroform-d) δ -109.20 (ddd, J = 15.0, 8.8, 6.2 Hz), -112.03 (t, J = 7.0 Hz). LCMS-EST (m/z): [M+H]+calculated for C22H20F3N3O4: 447.14; found: 448.32.
Example 100
198
Préparation of Compound 100 (lR,4R,12aS)-N-(2,4-difluorobenzyl)-7-hydroxy-6,8-dioxo-l,2,3,4,6,8,12,12aoctahydro-1,4-ethanodipyrido[ 1,2-a: 1 ',2'-d]pyrazine-9-carboxamide
100-A
100
Borane
THF
100-B
Hydrazine
inr, r.t. Boc
100-C
NaHCO3
EtOH/HzO
Boc
100-D
100
Step 1
A 100-mL rbf was charged with 100-A (2.0 g, 7.8 mmol) in THF (20 mL).
199
The reaction mixture was cooled to 0 °C. Borane dimethyl sulfide (2 N in THF, 17.6 mL) was slowly added in. Then the reaction mixture was stirred at room température for ovemight. The reaction mixture was cooled back to 0 °C. Methanol (8 mL) was added drop wise to quench the reaction. After concentration, the residue was purified by Combi Flash (40 g column, cartridge used) using hexanes - EA as eluents to afford 100-B. LCMS-ESI+ (m/z): [M+H]+ found: 242.
Step 2
A 100-mL rbf was charged with 100-B (1.8 g, 7.4 mmol), triphenylphosphine (4.3 g, 16.2 mmol) and phthalimide (1.8 g, 12.2 mmol) in THF (30 mL). Then the reaction mixture was cooled to 0 °C with stirring. DIAD (3.2 mL, 16.2 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room température for ovemight. After concentration, the residue was purified by Combi Flash (80 g column, cartridge used) using hexanes - EA as eluents to afford 100-C. LCMS-ESI+ (m/z): [M+H]+ found: 371.
Step 3
To a solution of 100-C (2.5 g, 6.8 mmol) in EtOH (50 mL) was added hydrazine monohydrate (1.7 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford
100-D. LCMS-ESE (m/z): [M+H]+found: 241.
Step 4
A 100-mL rbf was charged with 100-D (1.6 g, 6.7 mmol) and 100-E (2.3 g, 6.7 mmol) in éthanol (30 mL). Sodium bicarbonate (1.2 g, 1.4 mmol) in water (30 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for ovemight. The mixture was diluted with EA (200 mL) and washed with water (2 x). The aqueous fractions were extracted with EA (1 x), and the organic fractions were combined, dried (Na2SÛ4), and concentrated. The crude 100-F was used for next step without further purification. LCMS-ESI+ (m/z): [M+H]+found: 569.
200
Step 5
A 100-mL rbf was charged with 100-F (3.7 g, 6.5 mmol) in 4 N HCl /dioxane (38 mL). Then the reaction mixture was stirred at room température for 1 hour. After concentration, 3.2 g intermediate was obtained. The intermediate and DBU (5.1 g,
33.8 mmol) were dissolved in toluene (100 mL). The reaction mixture was heated to 110°C with stirring for 1 hour. After concentration, the residue was purified by Combi Flash (80 g column, cartridge used) using hexanes - EA as eluents to afford 100-G. LCMS-ESr (m/z): [M+H]+found: 423.
Step 6
A 100-mL rbf was charged with 100-G (2.0 g, 4.7 mmol) in THF (20 mL) and MeOH (20 mL). 1 N KOH (18.9 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for 1 hour. The reaction mixture was acidified by adding 1 N HCl (18.9 mL). After concentration, the residue was coevaporated with toluene (3 x). The crude acid (0.28 g, 0.72 mmol), 2, 4difluobenzylamine (0.2 g, 1.44 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.47 g, 3.6 mmol) and HATU (0.55 g, 1.44 mmol) were dissolved in DCM (20 mL). The reaction mixture was stirred at room température for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHCCh (2x), saturated NH4CI (2x) and dried over Na2SO4. After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 100-H. LCMS-ESI+ (m/z): [M+H]+ found: 520.
Step 7
A 50-mL rbf was charged with 100-H (0.36 g, 0.69 mmol) in TFA (5 mL). The reaction mixture was stirred at room température for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 100. ’H-NMR (400 MHz, Chloroform-d) δ 12.25 (m, IH), 10.47 (t, J = 5.9 Hz, IH), 8.30 (s, IH), 7.58 - 7.29 (m, IH), 6.98 - 6.50 (m, 2H), 4.62 (dd, J = 14.8, 4.9 Hz, 3H), 4.22 (t, J = 12.2 Hz, IH), 4.14 - 4.07 (m, IH), 3.96 (dd, J = 12.2, 3.1 Hz, IH), 2.26 - 1.44 (m, 9H). 19F-NMR (376 MHz, Chloroform-d) δ -112.38 (t, J = 7.7 Hz), -114.78 (q, J = 8.5 Hz). LCMS-ESI+ (m/z): found; 430.
201
Example 101
Préparation of Compound 101 (lR,4R,12aS)-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)-l,2,3,4,6,8,12,12aoctahydro-l,4-ethanodipyrido[l,2-a:r,2'-d]pyrazine-9-carboxamide
TFA
101
Step 1
A 100-mL rbf was charged with 101-A (0.3 g, 0.72 mmol) in THF (2 mL) and MeOH (2 mL). 1 N KOH (2.1 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for 1 hour. The reaction mixture was acidified by adding 1N HCl (2.1 mL). After concentration, the residue was co-evaporated with toluene (3 x). The crude acid (0.72 mmol), 2, 4, 6-trifluobenzylamine (0.23 g, 1.44 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.47 g, 3.6 mmol) and HATU (0.55 g, 1.44 mmol) were dissolved in DCM (20 mL). The reaction mixture was stirred at room température for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHCO3 (2x), saturated NH4CI (2x) and dried over Na2SO4. After
202 concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 101-B. LCMS-ESI+ (m/z): [M+H]+ found: 538.
Step 2
A 50-mL rbf was charged with 101-B (0.36 g, 0.67 mmol) in TFA (5 mL).
The reaction mixture was stirred at room température for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 101. 1H-NMR(400 MHz, Chloroform-d) δ 12.11 (s, IH), 10.40 (t, J =
5.8 Hz, IH), 8.28 (s, IH), 6.91 - 6.39 (m, 2H), 4.62 (ddd, J = 25.0, 6.5, 2.8 Hz, 3H), 4.21 (t, J =12.2 Hz, lH),4.09(dd, J = 12.5, 3.0 Hz, IH), 3.93 (dd, J = 12.2, 3.1 Hz, IH), 2.35
- 1.39 (m, 9H). 19F NMR (376 MHz, Chloroform-d) δ -112.38 (t, J = 7.7 Hz), -114.78 (q, J = 8.5 Hz). LCMS-EST (m/z): found: 448.
Example 102
Préparation of Compound 102 (lS,4S,12aR)-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)-l,2,3,4,6,8,12,12aoctahydro-l,4-ethanodipyrido[l,2-a:l',2'-d]pyrazine-9-carboxamide
102
203
Hydrazine
102-E
1) HCI/dioxane
2) DBU/Tol
102
Step 1
A 100-mL rbf was charged with 102-A (2.0 g, 7.8 mmol) in THF (20 mL).
The reaction mixture was cooled to 0 °C. Borane dimethyl sulfide (2 N in THF, 17.6 mL) was slowly added in. Then the reaction mixture was stirred at room température for ovemight. The reaction mixture was cooled back to 0 °C. Methanol (8 mL) was added drop wise to quench the reaction. After concentration, the residue was purified by Combi Flash (40 g column, cartridge used) using hexanes - EA as eluents to afford 102-B.
LCMS-ESI+ (m/z): [M+H]+ found: 242.
204
Step 2
A 100-mL rbf was charged with 102-B (1.8 g, 7.4 mmol), triphenylphosphine (4.3 g, 16.2 mmol) and phthalimide (1.8 g, 12.2 mmol) in THF (30 mL). Then the reaction mixture was cooled to 0 °C with stirring. DIAD (3.2 mL, 16.2 mmol) was slowly added to the reaction mixture. The reaction mixture was stirred at room température for ovemight. After concentration, the residue was purified by Combi Flash (80 g column, cartridge used) using hexanes - EA as eluents to afford 102-C. LCMS-ESI+ (m/z): [M+H]+ found: 371.
Step 3
To a solution of 102-C (2.5 g, 6.8 mmol) in EtOH (50 mL) was added hydrazine monohydrate (1.7 mL). The reaction mixture was heated to 70 °C with stirring for 3 hours. After filtration to remove the solid, the filtrate was concentrated to afford 102-D. LCMS-ESI+ (m/z): [M+H]+found: 241.
Step 4
A 100-mL rbf was charged with 102-D (1.6 g, 6.7 mmol) and 102-E (2.3 g, 6.7 mmol) in éthanol (30 mL). Sodium bicarbonate (1.2 g, 1.4 mmol) in water (30 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for ovemight. The mixture was diluted with EA (200 mL) and washed with water (2 x). The aqueous fractions were extracted with EA ( 1 x), and the organic fractions were combined, dried (NaîSCM), and concentrated. The crude 102-F was used for next step without fùrther purification. LCMS-ESH (m/z): [M+H]+ found: 569.
Step 5
A 100-mL rbf was charged with 102-F (3.7 g, 6.5 mmol) in 4 N HCl /dioxane (38 mL). Then the reaction mixture was stirred at room température for 1 hour. After concentration, 3.2 g intermediate was obtained. The intermediate and DBU (5.1 g,
33.8 mmol) were dissolved in toluene (100 mL). The reaction mixture was heated to 110°C with stirring for 1 hour. After concentration, the residue was purified by Combi Flash (80 g column, cartridge used) using hexanes - EA as eluents to afford 102-G.
205
LCMS-ESr (m/z): [M+H]*found: 423.
Step 6
A 100-mL rbf was charged with 102-G (0.3 g, 0.72 mmol) in THF (2 mL) and MeOH (2 mL). 1 N KOH (2.1 mL) was added to the reaction mixture. Then the reaction mixture was stirred at room température for 1 hour. The reaction mixture was acidified by adding 1N HCl (2.1 mL). After concentration, the residue was co-evaporated with toluene (3x). The crude acid (0.72 mmol), 2, 4, 6-trifluobenzylamine (0.23 g, 1.44 mmol), Ν,Ν-diisopropylethylamine (DIPEA) (0.47 g, 3.6 mmol) and HATU (0.55 g, 1.44 mmol) were dissolved in DCM (20 mL). The reaction mixture was stirred at room température for 2 hours. The mixture was diluted with EA (100 mL) and washed with saturated NaHCOs (2x), saturated NH4CI (2x) and dried over NaîSCU After concentration, the crude was purified by column chromatography on silica gel with hexane-EtOAc to afford 102-H LCMS-ESI+ (m/z): [M+H]+found: 538.
Step 7
A 50-mL rbf was charged with 102-H (0.36 g, 0.67 mmol) in TFA (5 mL). The reaction mixture was stirred at room température for 30 minutes. After concentration, the crude was purified by column chromatography on silica gel with EtOAc-MeOH to afford compound 102. Ή-NMR (400 MHz, Chloroform-d) δ 12.13 (s, IH), 10.40 (t, J =
5.8 Hz, IH), 8.28 (s, IH), 6.64 (t, J = 8.1 Hz, 2H), 4.89 - 4.41 (m, 3H), 4.22 (t, J = 12.2 Hz, IH), 4.09 (dd, J = 12.3, 3.1 Hz, IH), 3.95 (dd, J = 12.1, 4.1 Hz, IH), 2.45 - 1.60 (m, 9H). 19F-NMR(376 MHz, Chloroform-d) δ - 109.26 (ddd, J = 15.1, 8.8, 6.3 Hz), -111.99 (t, J = 6.9 Hz). LCMS-ESr (m/z): found: 448.
Example 103
Préparation of Compound 103 (lR,4R,12aR)-2,3-difluoro-7-hydroxy-6,8-dioxo-N-(2,4,6-trifluorobenzyl)1,2,3,4,6,8,12,12a-octahydro-l,4-methanodipyrido[l,2-a: l',2'-d]pyrazine-9carboxamide
206
O OH
103
DAST
DCM, -78 °C
2) Boc anyhdride
DIPEA, 2-MeTHF
1)Pd(OH)2, H2
HCl, éthanol
103-B
LiAIH4
THF, 0 °C
H
103-C
1)DIAD, PPh3 phthalimide THF
H
103-D
2) hydrazine hydrate EtOH
MeOH, 40 °C 103-E 103
Step 1
A solution of (lR,3R,4R,5R,6S)-methyl 5,6-dihydroxy-2-((S)-lphenylethyl)-2-azabicyclo[2.2.1]heptane-3-carboxylate (2.0g, 6.9mmol) in DCM (27mL) was cooled to -78 °C in a dry ice/acetone bath. To this solution was added DAST 10 (2.18 ml, 16.48 mmol) via plastic tipped pipette. The solution was stirred at -78 °C for minutes after which time it was removed from the bath, let warm slowly to room température, and stirred at room température for one hour. The reaction was quenched by slow addition of the reaction mixture to a stirring solution of saturated sodium bicarbonate (150mL) via plastic tipped pipette. The layers were separated and the
207 aqueous layer was back-extracted with dichloromethane. The combined organic layers were dried over magnésium sulfate, filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (7-28% ethyl acetate/hexane) to provide 103-A. 1H-NMR (400 MHz, Chloroform-<7) δ 7.43 - 7.16 (m, 5H), 5.01 - 4.60 (m, 2H), 3.85 (q, J= 7.1, 6.6 Hz, IH), 3.55 (s, 2H), 3.53 - 3.42 (m, 2H), 2.76 (dq, J = 5.1,2.0 Hz, IH), 2.19-2.07 (m, IH), 2.03 - 1.88 (m, IH), 1.39 (d, J= 6.7 Hz, 3H).
Steps 2 and 3
To a solution of 103-A (0.96 g, 3.24 mmol) in Ethanol (36.01 ml) and 1.25M HCl-ethanol (4.09 ml) was added 20% PdOH/C (1.14 g, 1.62 mmol) the suspension was stirred under an atmosphère of hydrogen for 22 hours. After filtering through Celite, the cake was washed with EtOH, the filtrate was concentrated under vacuum to dryness to afford the crude deprotected product which was assumed to be 3.24mmol for next step. LCMS-ESI+ (m/z): [M+H]+ calculated for C8Hi2F2NO2: 192.08; found: 192.110.
To the crude residue (0.62 g, 3.24 mmol) and Di-tert-butyl dicarbonate (1.06 g, 4.86 mmol) in 2-Methyltetrahydrofuran (32.43 ml) was added N,Ndiisopropylethylamine (0.56 ml, 0 mol). Upon completion, the reaction mixture was diluted with water, extracted into EtOAC (2x) and the organic fractions were washed with water, combined, dried (Na2SO4), and concentrated. The residue was purified by silica column chromatography (0-55% EtOAc/Hexanes) to afford 103-B. ^-NMR (400 MHz, Chloroform-d) δ 5.12-5.01 (m, IH), 4.92 (s, 1 H), 4.49 (s, IH), 4.14(d, J= 14.7 Hz, IH), 3.75 (s, 3H), 2.91 (s, IH), 2.24 - 1.98 (m, 2H), 1.47 (s, 5H), 1.38 (s, 5H). LCMS-ESU (m/z): [M+H]+ calculated for Ci3H2oF2N04: 292.13; found: 291.75.
Step 4
A solution of 103-B (0.68 g, 2.33 mmol) in THF (15 ml) was stirred in an ice bath as 1.0 M LiBH4 in THF (4.65 ml) was added and the resulting mixture was stirred at 0 °C for 30 minutes at which time it was shown to be complété by TLC. The reaction mixture was carefully treated with water (0.3 mL), then with NaOH (-15%, 3.5M, 0.3 mL), then finally with additional water (0.9 mL). The mixture was stirred at room
208 température for 15 minutes, and the ppt that formed was filtered, washed with diethyl ether and the supernate was concentrated to afford 103-C. ’H-NMR (400 MHz, Chloroform-d) δ 4.83 (s, IH), 4.56 (d, J = 10.5 Hz, IH), 4.37 (s, IH), 3.78 - 3.47 (m, 3H), 2.76 (s, IH), 2.36 - 2.18 (m, IH), 2.17 -1.98 (m, IH), 1.55 (s, IH), 1.48 (s, 9H).
Steps 5 and 6
A mixture of 103-C (0.59 g, 2.25 mmol), phthalimide (0.53 g, 3.6 mmol) and triphenylphosphine (1.3 g, 4.95 mmol) in THF (11 ml) was cooled in an ice bath. Diisopropyl Azodicarboxylate (0.97 ml, 4.95 mmol) was added. The mixture was then warmed up to room température and stirred for 14h and then concentrated in vacuo. The residue was dissolved in ether, stirred for 1 h, then the solids were filtered off and the fîltrate was concentrated. The residue was purified by silica column chromatography (10-31-91% EtOAc/hexanes) to afford the protected amino compound (assumed 2.25mmol of product). LCMS-ESI+ (m/z): [M+H]+ calculated for C20H23F2N2O4: 393.15; found: 392.77.
A solution of the protected amino compound (0.88 g, 2.25 mmol) and hydrazine hydrate (0.46 ml, 9.52 mmol) in éthanol (22 ml) was stirred at 60 °C for 2 h The reaction mixture was cooled in an ice bath, ether (10 ml) was added and the mixture was stirred for 30 min. The solid formed was filtered off and the fîltrate was concentrated under vacuum to dryness to give 103-D. ’H-NMR (400 MHz, Chloroformd) δ 5.17 - 4.61 (m, 2H), 4.37 (s, IH), 3.80 (s, IH), 3.11 - 2.77 (m, IH), 2.01 (s, 2H), 1.87 (s, IH), 1.83 (d, J = 7.4 Hz, IH), 1.46 (s, 9H), 1.30 (d, J = 6.4 Hz, IH), 1.27 (d, J = 6.3 Hz, 3H). LCMS-ESI+ (m/z): [M+H]+ calculated for C12H20F2N2O2: 263.15; found: 262.86.
Steps 7, 8 and 9
Compound 103 was prepared in a similar manner to compound 60 using 103-D in place of 41-E and using (2,4,6-trifluorophenyl)methanamine in place of (2,3dichlorophenyl)methanamine. A single diastereomer resulted. The stereochemistry of the fluorines is unknown. 1H-NMR (400 MHz, Chloroform-d) δ 8.08 (s, IH), 6.46 - 6.27 (m, 2H), 4.95 (d, J = 53.5 Hz, IH), 4.65 (d, J = 54.9 Hz, IH), 4.45 (s, IH), 4.33 (d, J =
209
5.6 Hz, 2H), 3.84 (t, J = 3.6 Hz, 2H), 2.75 (s, IH), 2.28 (p, J = 1.9 Hz, 2H), 2.20 (s, IH), 1.91 (dd, J = 33.3, 15.2 Hz, IH), 0.95 (s, IH). LCMS-ESI+ (m/z): [M+H]+ calculated for C21H17F5N3O4: 470.11; found: .470.13.
Antiviral Assay
Example 104
Antiviral Assays in MT4 Cells
For the antiviral assay utilizing MT4 cells, 0.4 pL of 189X test concentration of 3-fold serially diluted compound in DMSO was added to 40 pL of cell growth medium (RPMI 1640, 10% FBS, 1% penicilline/Streptomycine, 1% LGlutamine, 1% HEPES) in each well of 384-well assay plates (10 concentrations) in quidruplicate.
mL aliquots of 2 χ ΙΟ6 MT4 cells are pre-infected for 1 and 3 hours respectively at 37 °C with 25 pL (MT4) or of either cell growth medium (mock-infected) or a ffesh 1:250 dilution of an HIV-IIIb concentrated ABI stock (0.004 m.o.i. for MT4 cells). Infected and uninfected cells are diluted in cell growth medium and 35 pL of 2000 (for MT4) cells is added to each well of the assay plates.
Assay plates were then incubated in a 37 °C incubator. After 5 days of incubation, 25 pL of 2X concentrated CellTiter-Glo™ Reagent (catalog # G7573, Promega Biosciences, Inc., Madison, WI) was added to each well of the assay plate. Cell lysis was carried out by incubating at room température for 2-3 minutes, and then chemiluminescence was read using the Envision reader (PerkinElmer).
Compounds of the présent invention demonstrate antiviral activity in this assay as depicted in Table 1 below. Accordingly, the compounds of the invention may be useful for treating the prolifération ofthe HIV virus, treating AIDS, or delaying the onset of AIDS or ARC symptoms.
Table 1
210
Compound Number nM in MT-4
EC50 CCso
1 2.6 5819
2 2.2 3111
O 3 2.0 38446
4 14.8 45769
5 8.1 10452
6 5.3 53192
7 3.5 15610
8 2.5 13948
9 5.1 13451
10 6.1 3670
11 4.9 10274
12 5.9 3337
13 46.0 12666
14 65.5 4939
15 2.2 16268
16 1.5 13633
17 5.9 6613
18 4.1 10263
19 2.8 38690
20 o 3.3 27990
21 38.3 13010
22 64.3 4433
23 2.3 13444
24 6.1 12074
25 26.2 5233
26 10.3 8836
27 4.4 8751
28 15.6 18687
211
Compound Number nM in MT-4
ec50 CC50
29 13.9 9446
30 4.0 6828
31 9.0 4525
32 14.0 4684
33 43.5 3971
34 422.1 3585
35 157.0 15546
36 7.6 11424
37 10.2 19486
38 1.7 10223
39 3.6 12174
40 2.4 9560
41 2.1 15675
42 2.5 3544
43 6.9 10321
44 2.3 9869
45 2.4 15765
46 2.6 19295
47 1.9 11301
48 2.7 13967
49 o o J J. J 52219
50/51 (racemic mixture) 1.9 37173
52 15.0 12943
53 14.3 3347
54 15.6 3236
55 1.5 11100
56 3.1 17238
212
Compound Number nM in MT-4
EC50 CC50
57 2.3 11751
58 1.5 7694
59 3.1 22200
60 2.1 3308
61 1.8 25881
62 9.2 3492
63 2.5 3164
64 3.5 3332
65 2.4 2508
66 9.4 11848
67 10.7 2981
68 2.7 4175
69 1.9 4767
70 5.1 8413
71 2.6 4660
72 4.3 6255
73 1.8 9194
74 29.3 4340
75 2.8 5292
76 17.8 34581
77 5.6 10145
78 5.6 3198
79 3.4 12092
80 4.6 5045
81 1.9 12298
82 2.9 30434
83 1.9 27501
84 2.9 9727
213
Compound Number nM in MT-4
ECso cc50
85 2.0 10378
86 2.3 22405
88 2.9 3230
89 8.4 4629
90 5.7 8086
91 5.0 7183
92 18.6 4553
93 2.2 6158
94 11.5 51173
96 2.6 26586
97 2.1 17341
98 2.4 17947
99 2.0 8475
100 2.2 11580
101 2.1 11585
102 2.3 12042
103 10.3 35127
Example 105
Human PXR Activation Assay
Luciferase Reporter Gene Assay. A stably transformed tumor cell line (DPX2) was plated on 96-well microtiter plates. DPX2 cells harbor the human PXR gene 5 (NR1I2) and a luciferase reporter gene linked to two promoters identified in the human
CYP3 A4 gene, namely XREM and PXRE. The cells were treated with six concentrations of each compound (0.15 ~ 50 μΜ) and incubated for 24 hours. The number of viable cells was determined and the reporter gene activity was assessed. Positive control: Rifampicin at 6 concentrations (0.1 ~ 20 μΜ). %Emax relative to the maximum fold induction by 10 10 or 20 μΜ RBF was calculated for test compounds according to the following équation
214 which adjusts for the DMSO background: %Emas = (Fold induction - l)/(Maximum fold induction by RIF - 1) x 100%.
Table 2
Compound %Emax Ht
Number 15 μΜ
2 4.5
-> J 7.5
4 3
5 32
6 0
7 6
8 7
9 7
10 19
15 20
16 17
17 7
18 4
19 2
20 2
23 45
28 6
29 3
32 14
33 17
36 3
37 2
38 7
39 6
215
Compound %Emax at
Number 15 μΜ
40 0
41 11.5
42 21
43 18
44 4
45 19
46 34
47 11
48 5
54 2
55 24
56 O 3
57 3
58 1
59 4
60 3
61 1
63 13
64 8
66 0
67 0
68 6
69 5
70 10
71 o 3
72 4
73 7
75 0
216
Compound %Emax at
Number 15 μΜ
77 11
79 0
80 2
81 1
82 1
83 1
84 21
85 77
86 30
88 27
89 5
90 11
91 3
92 3
93 9
96 11
97 9
98 0
99 17
100 45
102 123
103 0
Example 106
OCT2 Inhibition Assay
The dose dépendent inhibition of OCT2 mediated uptake of a model substrate 14C-Tetraethylammonium (TEA) by test compounds was studied in wild-type 5 and OCT2-transfected MDCKII cells at 7 concentrations from 0.014 μΜ to 10 μΜ.
217
MDCKII cells were maintained in minimal essential medium (MEM) with l% Pen/Strep, 10% fêtai bovine sérum, and 0.25 mg/mL hygromycin B in an incubator set at 37 °C, 90% humidity and 5% CO2. 24 hours prior to assay, media containing 5 mM sodium butyrate were added to MDCKII cells in flasks, and cells were grown to 80-90% confluence. On assay day, cells were trypsinized and resuspended in Krebs-Henseleit Buffer (KHB), pH 7.4 at 5 x 106 million cells/mL. Cells were preincubated for 15 min in assay plate before addition of test compound or substrate.
Test compounds were serially diluted in DMSO and then spiked (2 pL) into in 0.4 mL KHB buffer containing wild-type or OCT2-transfected cells and incubated for 10 minutes. Assay was initiated with the addition of 0.1 mL of 100 μΜ 14C-TEA in KHB buffer (20 μΜ final concentration after mixing). The concentration of TEA is based on the Km. After 10 minutes of incubation, the assay mixture was quenched with addition of 0.5 mL of ice-cold IX PBS buffer. Samples were then centrifuged at 1000 rpm for 5 min and supernatants were removed. Wash steps were repeated four times with ice-cold PBS. Finally, the cell pellets were lysed with 0.2NNaOH and let sit at room température for at least 30 min to ensure complété lysis. Samples were then counted on liquid scintillation counter and dpm counts were used to perform the following calculations. The % inhibition was calculated as follows: % inhibition = [1- { [OCT2]i - [WT]ni} / {[OCT2]ni-[WT]ni}]*100 where, [OCT2]irepresents the dpm count in the presence oftest compound for either OCT2 cells, [OCT2]„i represents the dpm count in the absence of test compound for OCT2 cells and [WT]m represents the dpm count in the absence of test compound for wild type cells, respectively.
Table 3
Compound Number IC50 (nM)
2 240
3 250
5 2230
11 10000
218
Compound Number IC5o (nM)
13 610
36 10000
39 358
40 204
41 2823
42 487
45 137
47 6200
48 4909
55 476
63 42
64 94
77 3830
82 10000
83 10000
96 1357
98 3726
99 1506
100 450
The data in Tables 1,2 and 3 represent an average over time of each assays for each compound. For certain compounds, multiple assays hâve been conducted over the life of the project. Thus, the data reported in Tables 1, 2 and 3 include the data reported 5 in the priority documents, as well as data from assays run in the intervening period.
219
Example 107
Pharmacokinetic Analysis Following Oral or Intravenous Administration to Beagle Dogs
Pharmacokinetic analysis was performed on various test compounds following intravenous or oral administration to beagle dogs.
For pharmacokinetic analysis of intravenously administered compounds, the test compounds were formulated in 5% Ethanol, 55% PEG 300, and 40% water at 0.1 mg/mL for IV infusion. For pharmacokinetic analysis oforally administered compounds, the test compounds were formulated as an aqueous suspension in 0.1% Tween 20, 0.5% HPMC LV100 in Di Water at 1 mg/kg.
Each dosing group consisted of 3 male, non-naïve purebred beagle dogs. At dosing, the animais weighed between 10 to 13 kg. The animais were fasted ovemight prior to dose administration and up to 4 hr after dosing. For studies of intravenous administration, the test article was administered to the animais by intravenous infusion over 30 min. The rate of infusion was adjusted according to the body weight of each animal to deliver a dose of 0.5 mg/kg. For studies of oral administration, the test article was administered according to the body weight of each animal to deliver a dose of 1 mg/kg.
For pharmacokinetic analysis of intravenously administered compounds, serial venous blood samples (approximately 1 mL each) were taken from each animal at 0, 0.250, 0.483, 0.583, 0.750, 1.00, 1.50, 2.00, 4.00, 8.00, 12.0, and 24.0 hours after dosing. The blood samples were collected into Vacutainer™ tubes containing EDTAK2 as the anti-coagulant and were immediately placed on wet ice pending centrifugation for plasma. An LC/MS/MS method was used to measure the concentration of the test compound in plasma. An aliquot of 100 pL of each plasma sample was added to a clean 96 well plate, and 400 pL of cold acetonitrile/internal standard solution (ACN)/(ISTD) was added. After protein précipitation, an aliquot of 110 pL of the supernatant was transferred to a clean 96-well plate and diluted with 300 pL of water. An aliquot of 25 pL of the above solution was injected into a TSQ Quantum Ultra LC/MS/MS System utilizing a Hypersil Gold Cis HPLC column (50 X 3.0 mm, 5 pm; Thermo-Hypersil Part # 25105-053030). An Agilent 1200 sériés binary pump (P/N G1312A Bin Pump)
220 was used for elution and séparation, and an HT S Pal autosampler (LE AP Technologies, Carrboro, NC) was used for sample injection. A TSQ Quantum Ultra triple quadrupole mass spectrometer was utilized in sélective reaction monitoring mode (Thermo Finnigan, San José, CA). Liquid chromatography was performed using two mobile phases: mobile phase A contained 1% acetonitrile in 2.5 mM ammonium formate aqueous solution with pH of 3.0, and mobile phase B contained 90% acetonitrile in 10 mM ammonium formate with pH of 4.6. Non-compartmental pharmacokinetic analysis was performed on the plasma concentration-time data. The resulting data are shown in the first three columns of Table 4. In Table 4, CL refers to clearance, which characterizes the rate at which drug is removed from plasma. The lower the clearance of a drug is, the longer the élimination half-life is in the body. Vss refers to the steady state volume of distribution and indicates how well a drug is distributed into the tissues. The larger the Vss is, the longer the élimination half-life is in the body. MRT refers to mean résidence time, which is a measure of the average time molécules exist in the body.
For pharmacokinetic analysis of orally administered compounds, serial venous blood samples (approximately 0.3 mL each) were taken from each animal at time points of 0, 0.25, 0.50, 1.0, 2.0, 4.0, 6.0, 8.0, 12.0 and 24.0 hours after dosing. Blood samples were collected, prepared and analyzed in a similar way to the intranveous studies described above. Non-compartmental pharmacokinetic analysis was performed on the plasma concentration-time data. The resulting data are shown in the last three columns of Table 4. In Table 4, F (%) refers to oral bioavailability. Cmax refers to the peak plasma concentration of the compound after administration. AUC refers to area under the curve and is a measure of total plasma exposure of the indicated compound.
Table 4
Compo und # CL (L/h/kg) Vss (L/kg) MRT (h) F (%) aqueous suspension Cmax (jlM) aqueous suspension AUC (pM*h) aqueous suspension
98 0.047 0.16 3.3 n/a n/a n/a
83 0.161 0.38 2.4 n/a n/a n/a
221
55 0.058 0.24 4.2 n/a n/a n/a
77 0.300 0.64 2.2 n/a n/a n/a
41 0.015 0.11 7.5 10.7 2.4 16.3
42 0.020 0.15 7.1 28.0 4.5 28.6
47 0.014 0.10 7.4 12.6 2.8 20.4
8 0.498 0.87 1.8 n/a n/a n/a
7 0.510 1.20 2.3 n/a n/a n/a
o 3 0.047 0.23 4.9 18.7 1.2 9.2
2 0.030 0.20 6.5 40.7 7.8 66.1
Ail of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this spécification are incorporated herein by reference, in their entirety to 5 the extent not inconsistent with the présent description.
From the foregoing it will be appreciated that, although spécifie embodiments of the invention hâve been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.
222

Claims (108)

  1. What is claimed is:
    1. A compound having the following Formula (I):
    or a stereoisomer or pharmaceutically acceptable sait thereof, wherein:
    X is -O- or -NZ3- or -CHZ3-;
    W is -CHZ2-;
    Z1, Z2 and Z3 are each, independently, hydrogen or Ci-3alkyl, or wherein Z1 and Z2 or Z1 and Z3, taken together, form -L- wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, or -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, wherein at least one of Z1 and Z2 or Z1 and Z3, taken together, form -L-;
    Z4 is a bond, -CH2-, or -CH2CH2-;
    Y1 and Y2 are each, independently, hydrogen, Ci-3alkyl or Ci-3haloalkyl;
    R1 is phenyl substituted with one to three halogens; and each Ra is, independently, hydrogen, halo, hydroxyl or Ci-4alkyl.
  2. 2. A compound of claim 1 having the following Formula (II-A):
    223
    O OH (II-A)
  3. 3. A compound of claim 1 having the following Formula (II-B):
    O
    O OH (II-B)
  4. 4. A compound of claim 1 having the following Formula (II-C):
    (II-C)
  5. 5. A compound of any one of claims 1-4 wherein L is -C(Ra)2-.
  6. 6. A compound of any one of claims 1-4 wherein L is
    -C(Ra)2C(Ra)2-.
    224
  7. 7. A compound of any one of claims 1-4 wherein L is -C(Ra)2C(Ra)2C(Ra)2-,
  8. 8. A compound of any one of claims 1-7 wherein each Ra is hydrogen.
  9. 9. A compound of any one of claims 1-7 wherein one Ra is methyl and each remaining Ra is hydrogen.
  10. 10. A compound of any one of claims 1-7 wherein at least one Ra is halogen and each remaining Ra is hydrogen.
  11. 11. A compound of any one of claims 1 -7 wherein two Ra are halogen and each remaining Ra is hydrogen.
  12. 12. A compound of any one of claims 1-7 wherein one Ra is halogen and each remaining Ra is hydrogen.
  13. 13. A compound of any one of claims 1-2 or 5-12 wherein X is -O-.
  14. 14. A compound of any one of claims 1-2 or 5-12 wherein X is -NZ3-.
  15. 15. A compound of any one of claims 1-2 or 5-12 wherein X is -NH-.
  16. 16. A compound of any one of claims 1-2 or 5-12 wherein X is -CHZ3- and Z1 and Z3, taken together, form -L-.
  17. 17. A compound of claim 16 wherein Z2 is hydrogen.
  18. 18. A compound of any one of claims 1-2 or 5-12 wherein X is -CH2-,
    225
  19. 19. A compound of any one of claims 1 or 5-18 wherein Z4 is a bond or -CH2-,
  20. 20. A compound of any one of claims 1 or 5-18 wherein Z4 is -CH2-.
  21. 21. A compound of any one of claims 1 or 5-18 wherein Z4 is a bond.
  22. 22. A compound of any one of claims 1 or 5-21 wherein Y1 and Y2 are each independently hydrogen, methyl or trifluoromethyi.
  23. 23. A compound of any one of claims 1-22 wherein R1 is substituted with one halogen.
  24. 24. A compound of claim 23 wherein R1 is 4-fluorophenyl or 2fluorophenyl.
  25. 25. A compound of any one of claims 1-22 wherein R1 is substituted with two halogens.
  26. 26. A compound of claim 25 wherein R1 is 2,4-difluorophenyl, 2,3difluorophenyl, 2,6-difluorophenyl, 3-fluoro-4-chlorophenyl, 3,4-difluorophenyl, 2fluoro-4-chlorophenyl, or 3,5-difluorophenyl.
  27. 27. A compound of claim 26 wherein R1 is 2,4-difluorophenyl.
  28. 28. A compound of any one of claims 1-22 wherein R1 is substituted with three halogens.
  29. 29. A compound of claim 28 wherein R1 is 2,4,6-trifluorophenyl or 2,3,4-trifluorophenyl.
  30. 30. A compound of claim 29 wherein R1 is 2,4,6-trifluorophenyl.
    226
  31. 31. A compound of claim 1 selected from:
    227
    228
    F
    F
    F
    O OH
    F
    F
    F
    229
    O OH
    230
    231
    F
    232
    233
    234
    235
    O OH
    Ο OH
    236
    F
  32. 32. A compound of claim 1 selected from:
    237 and
    238
  33. 33. A pharmaceutical composition comprising a compound of any one of claims 1-32, or a stereoisomer or pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
  34. 34. A pharmaceutical composition of claim 33 further comprising one or more additional therapeutic agents.
  35. 35. A pharmaceutical composition of claim 34 wherein the one or more one additional therapeutic agents is an anti-HIV agent.
  36. 36. A pharmaceutical composition of claim 35 wherein the one or more additional therapeutic agents is selected from the group consisting ofHIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucléotide inhibitors of reverse transcriptase, and combinations thereof.
  37. 37. A method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of claims 1-32 or a pharmaceutical composition of claim 33.
  38. 38. A method of claim 37 fùrther comprising administering to the human a therapeutically effective amount of one or more additional therapeutic agents.
  39. 39. A method of claim 38 wherein the one or more additional therapeutic agents is an anti-HIV agent.
  40. 40. A method of claim 39 wherein the one or more additional therapeutic agents is selected from the group consisting ofHIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucléotide inhibitors of reverse transcriptase, and combinations thereof.
    239
  41. 41. Use of a compound of any one of claims 1-32 or a pharmaceutical composition of claim 33 for the treatment of an HIV infection in a human having or at risk of having the infection.
  42. 42. A use of claim 41 further comprising administering to the human a therapeutically effective amount of one or more additional therapeutic agents.
  43. 43. A use of claim 42 wherein the one or more additional therapeutic agents is an anti-HIV agent.
  44. 44. A use of claim 43 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease inhibitors, HIV nonnucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucléotide inhibitors of reverse transcriptase, and combinations thereof.
  45. 45. A compound as described in any one of claims 1-32, or a pharmaceutically acceptable sait thereof for use in medical therapy.
  46. 46. A compound as described in any one of claims 1-32, or a pharmaceutically acceptable sait thereof, for use in the therapeutic treatment of an HIV infection.
  47. 47. A compound having the following Formula (I):
    240 or a stereoisomer or pharmaceutically acceptable sait thereof, wherein:
    X is -O- or -NZ3- or -CHZ3-;
    W is -O- or -NZ2- or -CHZ2-;
    Z1, Z2 and Z3 are each, independently, hydrogen or Ci-3alkyl, or wherein Z1 and Z2 or Z1 and Z3, taken together, form -L- wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2 OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2, -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C( Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2C(R a)2-, -C(Ra)2C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SO2C(Ra)2-, -C (Ra)2SO2NRaC(Ra)2- or -C(Ra)2NRaSO2C(Ra)2-;
    Z4 is a bond or -CH2-, -CH2CH2-, -CH2CH2CH2-, -CH2OCH2-, -CH2NRaCH2-, -CH2SCH2-,-CH2S(O )CH2- or -CH2SO2CH2-;
    Y1 and Y2 are each, independently, hydrogen, Ci-3alkyl or Ci-ahaloalkyl, or Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms, wherein the carbocyclic or heterocyclic ring is optionally substituted with one or more Ra;
    R1 is optionally substituted aryl or optionally substituted heteroaryl; and
    241 each Ra is, independently, hydrogen, halo, hydroxyl or Ci-4alkyl, or wherein two Ra groups, together with the carbon atom to which they are attached, form C=O, and wherein at least one of: (i) Z1 and Z2 or Z1 and Z3, taken together, form -L-; or (ii) Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
  48. 48. A compound of claim 47 wherein W is -CHZ2-
  49. 49. A compound of claim 47 or 48 wherein Z1 and Z2 or Z1 and Z3, taken together, form -L-.
  50. 50. A compound of claim 49 having one of the following Formulas (II-A), (Π-B), or (II-C):
    O OH (II-A)
    O OH ; or (II-B)
    242
    Ο
    Ο ΟΗ (II-C) wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2 OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2, -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C( Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2C(R a)2-, -C(Ra)2C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SO2C(Ra)2-, -C (Ra)2SO2NRaC(Ra)2- or -C(Ra)2NRaSO2C(Ra)2-,
  51. 51. A compound of claim 47 or 48 wherein Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
  52. 52. A compound of claim 51 having one of the following Formulas (III-A), (III-B), (ΠΙ-C) or (III-D):
    243 (III-B) ; or (III-C) wherein Z1 and Z3 are each, independently, hydrogen or Ci-3alkyl.
  53. 53. A compound of claim 51 having one of the following Formulas (III-E), (ΠΙ-F), (III-G) or (ΙΠ-Η).
    244
    245 (ΠΙ-Η) wherein Z1 and Z3 are each, independently, hydrogen or Ci-3alkyl.
  54. 54. A compound of claim 47 or 48 wherein both (i) Z1 and Z2 or Z1 and Z3, taken together, form -L-, and (ii) Y1 and Y2, together with the carbon atom to which they are attached, form a carbocyclic ring having from 3 to 6 ring atoms or a heterocyclic ring having from 3 to 6 ring atoms.
  55. 55. A compound of claim 54 having one of the following Formulas (IV-AA), (IV-AB), (IV-AC), (IV-AD), (IV-AE), (IV-AF), (IV-AG) or (IV-AH):
    O OH (IV-AA)
    O OH (IV-AB)
    O OH
    246 (IV-AD)
    O OH (IV-AE)
    O OH (IV-AF)
    247
    O OH ; or wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2 OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2, -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C( Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2C(R a)2-, -C(Ra)2C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SO2C(Ra)2-, -C (Ra)2SO2NRaC(Ra)2- or -C(Ra)2NRaSO2C(Ra)2-,
  56. 56. A compound of claim 54 having one of the following Formulas (IV-BA), (IV-BB), (IV-BC), (IV-BD), (IV-BE), (IV-BF), (IV-BG) or (IV-BH):
    248 (IV-BA)
    O OH (IV-BB)
    O OH (IV-BD)
    249
    250 (IV-BH) wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2C(Ra)2C(Ra)2-, -C(Ra)2 OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2, -C(Ra)2OC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2C(Ra)2-, -C(Ra)2C( Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2C(R a)2-, -C(Ra)2C(Ra)2S(O)C(Ra)2-, -C(Ra)2SO2C(Ra)2C(Ra)2-, -C(Ra)2C(Ra)2SO2C(Ra)2-, -C (Ra)2SO2NRaC(Ra)2- or -C(Ra)2NRaSO2C(Ra)2-,
  57. 57. A compound of any one of claims 47-50 or 54-56 wherein L is -C(Ra)2-, -C(Ra)2C(Ra)2- -C(Ra)2C(Ra)2C(Ra)2-, or-C(Ra)2C(Ra)2C(Ra)2C(Ra)2-.
  58. 58. A compound of claim 57 wherein L is -C(Ra)2-.
  59. 59. A compound of claim 57 wherein L is -C(Ra)2C(Ra)2-.
  60. 60. A compound of claim 57 wherein L is -C(Ra)2C(Ra)2C(Ra)2-.
  61. 61. A compound of any one of claims 47-50 or 54-60 wherein each Ra is hydrogen.
  62. 62. A compound of any one of claims 47-50 or 54-60 wherein one Ra is methyl and each remaining Ra is hydrogen.
  63. 63. A compound of any one of claims 47-50 or 54-60 wherein at least one Ra is halogen and each remaining Ra is hydrogen.
  64. 64. A compound of any one of claims 47-50 or 54-60 wherein two Ra are halogen and each remaining Ra is hydrogen.
    251
  65. 65. A compound of any one of claims 47-50 or 54-60 wherein one Ra is halogen and each remaining Ra is hydrogen.
  66. 66. A compound of any one of claims 47-50 or 54-56 wherein L is -C(Ra)2OC(Ra)2-, -C(Ra)2NRaC(Ra)2-, -C(Ra)2SC(Ra)2-, -C(Ra)2S(O)C(Ra)2-, or -C(Ra)2SO2C(Ra)2-,
  67. 67. A compound of any one of claims 47-50 or 54-56 wherein L is -C(Ra)2OC(Ra)2-,
  68. 68. A compound of claim 66 or 67 wherein each Ra is hydrogen.
  69. 69. A compound of claim 66 or 67 wherein one Ra is methyl and each remaining Ra is hydrogen.
  70. 70. A compound of claim 66 or 67 wherein at least one Ra is halogen and each remaining Ra is hydrogen.
  71. 71. A compound of claim 66 or 67 wherein two Ra are halogen and each remaining Ra is hydrogen.
  72. 72. A compound of claim 66 or 67 wherein one Ra is halogen and each remaining Ra is hydrogen.
  73. 73. A compound of any one of claims 47-55 or 57-72 wherein X is -O-.
  74. 74. A compound of claim 73 wherein Z2 is hydrogen.
  75. 75. A compound of any one of claims 47-72 wherein X is -NZ3-.
  76. 76. A compound of any one of claims 47-72 wherein X is -NH-.
    252
  77. 77. A compound of any one of claims 47-55 or 57-72 wherein X is -CHZ3-
  78. 78. A compound of any one of claims 47-55 or 57-72 wherein X is -
    CH2-,
  79. 79. A compound of any one of claims 47-49, 51, 54 or 57-78 wherein
    Z4 is a bond or -CH2-.
  80. 80. A compound of any one of claims 47-49, 51, 54 or 57-78 wherein
    Z4 is -CH2-.
  81. 81. A compound of any one of claims 47-49, 51, 54 or 57-78 wherein Z4 is a bond.
  82. 82. A compound of any one of claims 47-49 or 57-81 wherein Y1 and Y2 are each independently hydrogen, methyl or trifluoromethyl.
  83. 83. A compound of any one of claims 47-82 wherein R1 is phenyl.
  84. 84. A compound of any one of claims 47-82 wherein R1 is pyridinyl.
  85. 85. A compound of any one of claims 47-84 wherein R1 is substituted with at least one halogen.
  86. 86. A compound of any one of claims 47-84 wherein R1 is substituted with one halogen.
  87. 87. A compound of claim 86 wherein R1 is 4-fluorophenyl or 2fluorophenyl.
    253
  88. 88. A compound of any one of claims 47-84 wherein R1 is substituted with two halogens.
  89. 89. A compound of claim 88 wherein R1 is 2,4-difluorophenyl, 2,3difluorophenyl, 2,6-difluorophenyl, 3-fluoro-4-chlorophenyl, 3,4-difluorophenyl, 2fluoro-4-chlorophenyl, or 3,5-difluorophenyl.
  90. 90. A compound of claim 89 wherein R1 is 2,4-difluorophenyl.
  91. 91. A compound of any one of claims 47-84 wherein R1 is substituted with three halogens.
  92. 92. A compound of claim 91 wherein R1 is 2,4,6-trifluorophenyl or 2,3,4-trifluorophenyl.
  93. 93. A compound of claim 92 wherein R1 is 2,4,6-trifluorophenyl.
  94. 94. A compound of claim 85 wherein R1 is 3-trifluoromethyl-4fluorophenyl or 2-cyclopropoxy-4-fluorophenyl.
  95. 95. A pharmaceutical composition comprising a compound of any one of claims 47-94, or a stereoisomer or pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable carrier, diluent or excipient.
  96. 96. A pharmaceutical composition of claim 95 further comprising one or more additional therapeutic agents.
  97. 97. A pharmaceutical composition of claim 96 wherein the one or more one additional therapeutic agents is an anti-HIV agent.
    254
  98. 98. A pharmaceutical composition of claim 97 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucléotide inhibitors of reverse transcriptase, and combinations thereof.
  99. 99. A method of treating an HIV infection in a human having or at risk of having the infection by administering to the human a therapeutically effective amount of a compound of any one of claims 47-94 or a pharmaceutical composition of claim 95.
  100. 100. A method of claim 99 further comprising administering to the human a therapeutically effective amount of one or more additional therapeutic agents.
  101. 101. A method of claim 100 wherein the one or more additional therapeutic agents is an anti-HIV agent.
  102. 102. A method of claim 101 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease inhibitors, HIV non-nucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucléotide inhibitors of reverse transcriptase, and other drugs for treating HIV, and combinations thereof.
  103. 103. Use of a compound of any one of claims 47-94 or a pharmaceutical composition of claim 95 for the treatment of an HIV infection in a human having or at risk of having the infection.
  104. 104. A use of claim 103 fùrther comprising administering to the human a therapeutically effective amount of one or more additional therapeutic agents.
  105. 105. A use of claim 104 wherein the one or more additional therapeutic agents is an anti-HIV agent.
    255
  106. 106. A use of claim 105 wherein the one or more additional therapeutic agents is selected from the group consisting of HIV protease inhibitors, HIV nonnucleoside inhibitors of reverse transcriptase, HIV nucleoside inhibitors of reverse transcriptase, HIV nucléotide inhibitors of reverse transcriptase, and other drugs for treating HIV, and combinations thereof.
  107. 107. A compound as described in any one of claims 47-94, or a pharmaceutically acceptable sait thereof for use in medical therapy.
  108. 108. A compound as described in any one of claims 47-94, or a pharmaceutically acceptable sait thereof, for use in the therapeutic treatment of an HIV infection.
    256
    ABSTRACT
    Compounds for use in the treatment of human immunodeficiency virus (HIV) infection are disclosed. The compounds hâve the following Formula (I):
    including stereoisomers and pharmaceutically acceptable salts thereof, wherein R1, X, W, Y1, Y2, Z1, and Z4 are as defined herein. Methods associated with préparation and use of such compounds, as well as pharmaceutical compositions comprising such compounds, are also disclosed.
OA1201500240 2012-12-21 2013-12-19 Polycyclic-carbamoylpyridone compounds and their pharmaceutical use. OA17631A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US61/745,375 2012-12-21
US61/788,397 2013-03-15
US61/845,803 2013-07-12

Publications (1)

Publication Number Publication Date
OA17631A true OA17631A (en) 2017-05-15

Family

ID=

Similar Documents

Publication Publication Date Title
AU2018232957B2 (en) Polycyclic-carbamoylpyridone compounds and their pharmaceutical use
OA17631A (en) Polycyclic-carbamoylpyridone compounds and their pharmaceutical use.