WO1997027180A1 - Aspartyl protease inhibitors - Google Patents

Aspartyl protease inhibitors Download PDF

Info

Publication number
WO1997027180A1
WO1997027180A1 PCT/US1997/001610 US9701610W WO9727180A1 WO 1997027180 A1 WO1997027180 A1 WO 1997027180A1 US 9701610 W US9701610 W US 9701610W WO 9727180 A1 WO9727180 A1 WO 9727180A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
group
formula
compound according
mmol
Prior art date
Application number
PCT/US1997/001610
Other languages
French (fr)
Inventor
Roger Dennis Tung
Francesco Gerald Salituro
David D. Deininger
Govinda Rao Bhisetti
Christopher Todd Baker
Andrew Spaltenstein
Wieslaw M. Kazmierski
Clarence Webster Andrews, Iii
Original Assignee
Vertex Pharmaceuticals Incorporated
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
Priority claimed from US08/592,777 external-priority patent/US5883252A/en
Application filed by Vertex Pharmaceuticals Incorporated filed Critical Vertex Pharmaceuticals Incorporated
Priority to EP97904911A priority Critical patent/EP0882022A1/en
Priority to EA199800661A priority patent/EA199800661A1/en
Priority to AU17580/97A priority patent/AU709239B2/en
Priority to JP9527124A priority patent/JP2000501111A/en
Priority to BR9707086A priority patent/BR9707086A/en
Priority to IL12547997A priority patent/IL125479A0/en
Publication of WO1997027180A1 publication Critical patent/WO1997027180A1/en
Priority to NO983435A priority patent/NO983435L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • C07D207/2632-Pyrrolidones with only hydrogen atoms or radicals containing only hydrogen and carbon atoms directly attached to other ring carbon atoms
    • C07D207/272-Pyrrolidones with only hydrogen atoms or radicals containing only hydrogen and carbon atoms directly attached to other ring carbon atoms with substituted hydrocarbon radicals directly attached to the ring nitrogen atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/04Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D233/28Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/30Oxygen or sulfur atoms
    • C07D233/32One oxygen atom
    • C07D233/36One oxygen atom with hydrocarbon radicals, substituted by nitrogen atoms, attached to ring nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/281,4-Oxazines; Hydrogenated 1,4-oxazines
    • C07D265/301,4-Oxazines; Hydrogenated 1,4-oxazines not condensed with other rings
    • C07D265/321,4-Oxazines; Hydrogenated 1,4-oxazines not condensed with other rings with oxygen atoms directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/281,4-Oxazines; Hydrogenated 1,4-oxazines
    • C07D265/341,4-Oxazines; Hydrogenated 1,4-oxazines condensed with carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • C07D285/01Five-membered rings
    • C07D285/02Thiadiazoles; Hydrogenated thiadiazoles
    • C07D285/04Thiadiazoles; Hydrogenated thiadiazoles not condensed with other rings
    • C07D285/101,2,5-Thiadiazoles; Hydrogenated 1,2,5-thiadiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems

Definitions

  • the present invention relates to a novel class of compounds which are aspartyl protease inhibitors.
  • this invention relates to a novel class of HIV aspartyl protease inhibitors cnaractenzed by specific structural and physicochemical features.
  • This invention also relates to pharmaceutical compositions comprising these compounds.
  • the compounds and pharmaceutical compositions of this invention are particularly well suited for inhibiting HIV-1 and HIV-2 protease activity ana consequently, may be advantageously used as anti- viral agents against the HIV-1 and HIV-2 viruses.
  • Tms invention also relates to methods for inhibiting aspartyl protease activity, methods for treating viral infections using the compounds and compositions of this invention, and methods for making intermediates and compounds of this invention.
  • the human immunodeficiency virus (“HIV”) is the causative agent for acquired immunodeficiency syndrome (“AIDS”) — a disease characterized by the destruction of the immune system, particularly of CD4
  • T-cells with attendant susceptibility to opportunistic infections — and its precursor AIDS-related complex (“ARC”) — a syndrome characterized by symptoms such as persistent generalized lymphadenopathy, fever and weight loss.
  • ARC AIDS-related complex
  • HIV encodes the production of a protease which carries out post-translational cleavage of precursor polypeptides in a process necessary for the formation of infectious vi ⁇ ons (S. Crawford et al., "A Deletion Mutation in the 5' Part of the pol Gene of Moloney Murine Leukemia Virus Blocks Proteolytic Processing of the gag and pol Polyprotems", J. Virol., 53, p. 899 (1985) ) .
  • These gene products include pol. which encodes the virion RNA-dependent DNA polymerase
  • a number of synthetic anti-viral agents have been designed to target various stages in the replication cycle of HIV. These agents include compounds which block viral binding to CD4 T- lymphocytes (for example, soluble CD4) , and compounds which interfere with viral replication by inhibiting viral reverse transcriptase (for example, didanosine and zidovudme (AZT) ) and inhibit integration of viral DNA into cellular DNA (M.S. Hirsh and R.T. D'Aqulia, "Therapy for Human Immunodeficiency Virus Infection", N.F.n ⁇ .J.Med. , 328, p. 1686 (1993)).
  • CD4 T- lymphocytes for example, soluble CD4
  • compounds which interfere with viral replication by inhibiting viral reverse transcriptase for example, didanosine and zidovudme (AZT)
  • AZT didanosine and zidovudme
  • agents which are directed primarily to early stages of viral replication, do not prevent the production of infectious virions in chronically infected cells. Furthermore, administration of some of these agents m effective amounts has led to cell-toxicity and unwanted side effects, such as anemia and bone marrow suppression.
  • the present invention provides a novel class of compounds, and pharmaceutically acceptable derivatives thereof, that are useful as inhibitors of aspartyl proteases, and m particular, HIV aspartyl protease.
  • the compounds of this invention can be used alone or in combination with other therapeutic or prophylactic agents, such as anti-virals, antibiotics, immunomodulators or vaccines, for the treatment or prophylaxis of viral infection.
  • the compounds of this invention are capable of inhibiting HIV viral replication in human CD cells including T- cells, monocytic lines including macrophages and dendrocytes and other permissive cells. These compounds are useful as therapeutic and prophylactic agents to treat or prevent infection by HIV-1 and related viruses which may result in asymptomatic infection, AIDS-related complex ("ARC"), acquired immunodeficiency syndrome (“AIDS”), or similar d sease of the immune system.
  • ARC AIDS-related complex
  • AIDS acquired immunodeficiency syndrome
  • each X and X' is independently selected from the group consisting of -C (O) - , -C (0) C (0) - , -S (0) - and
  • R C_-C 6 alkyl; C 2 -C 6 alkenyl; C 2 -C 6 alkynyl; C 3 -C 6 cycloalkyl optionally fused with R ; C 5 -C 6 cycloalkenyl optionally fused with R ; and where two R 's are
  • the R 's together with their attached geminal atom may form a spirocarbocyclic or spiroheterocyclic ring system
  • each R 3 is independently selected from oxo, OR9,
  • each R is independently selected from from the group consisting of OR 9 ; N(R 9 ) 2 ; X-R 9 ; C(0)N(R 9 ) 2 ; R 6 ;
  • R where any member of R may be optionally substituted by one or more groups independently
  • each R is independently selected from the group consisting of H, OH, 0 and R ; each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl may be optionally substituted with one or more groups selected
  • each R is independently selected from the group consisting of hydrogen, OH and 0;
  • p each R is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, and heterocyclyl;
  • each R is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heterocyclyl, aralkyl, carbocyclylalkyl and heterocyclylalkyl wherein any aryl, carbocyclyl or p heterocyclyl may be optionally fused with R and
  • each G is independently selected from the group
  • An alternate object of this invention is a novel class of compounds represented by formula IV:
  • X and X' are independently -C(O)- or -S(0) ? -,
  • Y is -(C(R 2 ) 2 )-M-, -(C(R 2 ) 2 ; -N(R 2 )- or -N(R 2 )
  • Another object of this invention is a novel class of compounds represented by formula V:
  • X is -C(O)- or -S(0) 2 -;
  • Y is -(C(R 2 ) 2 )-M-, -(C(R 2 ) 2 ) p -, -N(R 2 )- or -N(R 2 )-
  • R 10 is 0 or H 2 ; each R is independently H, OH or 0, wherein both
  • R ,11 are not simultaneously hydrogen;
  • Z is a structure of formula VI:
  • any structure of formula VI is optionally fused with an aryl, carbocyclic or heterocyclic ring and is optionally substituted with 1-3 substituents independently selected from R ; and each R 1, R2 R 8 , G, M, Q and X' is independently as defined for formula I - sa ⁇
  • lt is also an object of this invention to provide pharmaceutical compositions comprising the compounds of formulas I, IV and V and methods for their use as inhibitors of aspartyl protease, and particularly, HIV aspartyl protease.
  • -S0 - and “-S(0) 2 -” as used herein refer to a sulfone or sulfone derivative (i.e., both appended groups linked to the S), and not a sulfinate ester.
  • alkoxy refers to an alkyl ether radical, wherein the term “alkyl” is as defined above.
  • suitable alkyl ether radicals include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert- butoxy and the like.
  • alkyl refers to a straight-chain or branch-chain saturated aliphatic hydrocarbon radical containing the specified number of carbon atoms, or where no number is specified, preferably from 1-10 and more preferably from 1-5 carbon atoms.
  • alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec- butyl, tert-butyl, pentyl, isoamyl, n-hexyl and the like.
  • alkenyl refers to a straight-chain or branched-chain mono- or poly-unsaturated aliphatic hydrocarbon radical containing the specified number of carbon atoms, or where no number is specified, preferably from 2-10 carbon atoms and more preferably, from 2-6 carbon atoms.
  • alkenyl radicals include, but are not limited to, ethenyl, E- and Z-propenyl, isopropenyl, E- and Z-butenyl, E- and Z-isobutenyl, E- and Z-pentenyl, E- and Z-hexenyl, E,E-, E,Z-, Z,E- and Z,Z-hexadienyl and the like.
  • anti-viral agent refers to a compound or drug which possesses viral inhibitory activity.
  • agents include reverse transcriptase inhibitors (including nucleoside and non-nucleoside analogs) and protease inhibitors.
  • the protease inhibitor is an HIV protease inhibitor.
  • nucleoside analog reverse transcriptase inhibitors include, but are not limited to, zidovudme (AZT) , dideoxycytidme (ddC) , didanosme (ddl), stavudme (d4T) , 3TC, 935U83, 1592U89 and 524W91.
  • non-nucleoside analog reverse transcriptase inhibitor examples include, but are not limited to TIBO, delavirdme (U90) and nevirapme.
  • HIV protease inhibitors include, but are not limited to VX-478 (Vertex, also known as 141 94 (Glaxo-Wellcome) and KVX-478 (Kissei)), saqumavir (Ro 31-8959, Roche), mdmavir (L-735,524, Merck)), ⁇ tonavir (ABT 538,
  • aryl refers to a carbocyclic aromatic radical (such as phenyl or naphthyl) containing the specified number of carbon atoms, preferably from 6-14 carbon atoms, and more preferably from 6-10 carbor atoms.
  • aryl radicals include, but are not limited to phenyl, naphthyl, indenyl, mdanyl, azulenyl, fluorenyl, anthracenyl and the like.
  • carbocycle and “carbocyclyl” radical, refers to a non-aromatic stable 3- to 8- membered carbon ring which may be saturate ⁇ , ono- unsaturated or poly-unsaturated.
  • the carbocycle may be attached at any endocyclic carbon atom which results in a stable structure.
  • Preferred carbocycles have 5-6 carbons.
  • heterocycle and “heterocyclyl” radical, unless otherwise defined herein, refers to a stable 3-7 membered monocyclic heterocyclic ring or 8- 11 membered bicyclic heterocyclic ring which is either saturated or unsaturated, and which may be optionally benzofused if monocyclic.
  • Each heterocycle consists of one or more carbon atoms and from one to four heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur.
  • nitrogen and sulfur heteroatoms include any oxidized form of nitrogen and sulfur, and the quaternized form of any basic nitrogen.
  • any ring nitrogen may be optionally substituted with a substituent R , as defined herein for compounds of formula I .
  • a heterocyclyl radical may be attached at any endocyclic carbon or heteroatom which results in the creation of a stable structure.
  • Preferred heterocycles include 5-7 membered monocyclic heterocycles and 8-10 memebered bicyclic heterocycles.
  • Preferred heterocycles defined above include, for example, benzimidazolyl, imidazolyl, lmidazolinoyl, lmidazolidinyl, quinolyl, isoquinolyl, indolyl, mdazolyl, mdazolmolyl, perhydropyridazyl, pyridazyl, pyridyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazmyl, qumoxolyl, piperidmyl, pyranyl, pyrazolmyl, piperazmyl, pyrimidmyl, pyridazmyl, morpholinyl,
  • halogen refers to a radical of fluorine, chlorine, bromine or iodine.
  • HIV protease and “HIV aspartyl protease” are used interchangeably and refer to the aspartyl protease encoded by the human immunodeficiency virus type 1 or 2. In a preferred embodiment of this invention, these terms refer to the human immunodeficiency virus type 1 aspartyl protease.
  • inert solvent refers to a solvent reaction medium which allows the reagents to react together at a substantially increased rate relative to any reagent reacting with the designated solvent.
  • LG refers to groups readily displaceable by a nucleophile, such as an amine, alcohol, phosphorous or thiol nucleophile or their respective anions .
  • a nucleophile such as an amine, alcohol, phosphorous or thiol nucleophile or their respective anions .
  • Such leaving groups are well known and include carboxylates, N-hydroxysuccinimide, N-hydroxybenzotriazole, halogen (halides) , triflates, tosylates, mesylates, alkoxy, thioalkoxy, phosph ates, phosphonates and the like.
  • Other potential nucleophiles include organometallic reagents known to those skilled in the art.
  • protecting group refers to a suitable chemical group which may be attached to a functional group and removed at a later stage to reveal the intact functional group. Examples of suitable protecting groups for various functional groups are described in T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis. 2d. Ed.. John Wiley and Sons (1991) ; L. Fieser and M. Fieser, F eser and Fieser's Reagents for Organic Synthesis. John Wiley and Sons (1994) ; L. Paquette, ed. Encyclopedia of Reagents for Organic Synthesis. John Wiley and Sons (1995) .
  • fused refers to a structure wherein two distinct ring systems are joined together such that both rings share at least two common atoms. This can be envisioned as the replacement of a carbon-hydrogen or nitrogen-hydrogen bond on a ring atom with a carbon- carbon (from a second ring) or nitrogen-carbon (from a second ring) bond.
  • a cyclohexyl ring fused to a second cyclohexyl ring results in a decahydronaphthalene
  • a cyclohexyl ring fused to a piperidine ring results in a decahydroqumol e or decahydroisoqu olme
  • a phenyl ring fused to a thiazole ring results in a oenzothiazole.
  • substituents are those which enhance protease inhibitory activity or intracellular antiviral activity in permissive mammalian cells or immortalized mammalian cell lines, or which enhance deliverability - 1 6 -
  • pharmaceutically effective amount refers to an amount effective in treating HIV infection in a patient either as monotherapy or n combination with other agents.
  • treating refers to the alleviation of symptoms of a particu-ar disorder m a patient or the improvement of an ascertainable measurement associated with a particular disorder. Specifically, with respect to HIV, effective treatment using the compounds and compositions of tnis invention would result in an improvement in an HIV associated ascertainable measurement.
  • prolactically effective amount refers to an amount effective m preventing HIV infection in a patient.
  • patient refers to a mammal, including a human.
  • pharmaceutically acceptable carrier or adjuvant refers to a carrier or adjuvant that may be administered to a patient, together with a compoun ⁇ of this invention, and which does not destroy the pharmacological activity thereof and is nontoxic when administered doses sufficient to deliver a therapeutic amount of the antiretroviral agent.
  • the compounds of this invention, including the compounds of formula I are defined to include pharmaceutically acceptable ⁇ erivatives or prodrugs thereof.
  • a "pharmaceutically acceptable derivative or prodrug” means any pharmaceutically acceptable salt, ester, salt of an ester, or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing (directly or indirectly) a compound of this invention or an hibitorily active metabolite or residue thereof.
  • Particularly favore ⁇ derivatives and prodrugs are those that increase tne bioavailability of the compounds of this invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycollic, lactic, salicylic, succmic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene- 2-sulfon ⁇ c and benzenesulfonic acids.
  • Other acids, such as oxalic while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
  • Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N- (C;L_ 4 alkyl) 4 salts.
  • thiocaroamates refers to compounds containing the functional group N-S0 2 -0.
  • the compounds of this invention contain one or more asymmetric carbon atoms and thus occur as racemates and racemic mixtures, single enantiomers, diastereo eric mixtures and individual diastereomers. All such lsomeric forms of these compounds are expressly mcluded in the present invention.
  • Each stereogenic carbon may be of the R or S configuration.
  • stable refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a mammal or for use in affinity chromatography applications) .
  • such compounds are stable at a temperature of 40°C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • the compounds of the present invention may be used in the form of salts derived from inorganic or organic acids. Included among such acid salts, for example, are the following: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camph- orsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, gluconepta- noate, glycerophosphate, hemisulfate, heptanoate, hexa ⁇ noate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicot ate, oxalate, pamoate, pectinate, per
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein.
  • the basic nitrogen can be quaternized with any agents known to those of ordinary skill in the art including, for example, lower alkyl halides, such as methyl, ethyl, propyl and butyl chloride, bromides and iodides; dialkyl sulfates including dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aralkyl halides including benzyl and phenethyl bromides. Water or oil-soluble or dispersible products may be obtained by such quaternization.
  • R C 5 -C 6 cycloalkenyl optionally fused with R ; and where R 's are attached to adjacent atoms, the R 's together with their attached adjacent atoms form a carbocyclic or heterocyclic ring system which may be optionally fused with R ; where any member of R may be
  • each R is independently selected from hydrogen
  • the R 's together with their attached gemmal atom may form a spirocarbocyclic or spiroheterocyclic ring system
  • each R is independently selected from oxo, OR " ,
  • each R is independently selected from from tne
  • each R is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heterocyclyl, aralkyl, carbocyclylalkyl and heterocyclylalkyl wherein any aryl, carbocyclyl or p heterocyclyl may be optionally fused with R and p wherein any member of R may be optionally substituted by one or more groups independently selected from the
  • 8 8 P group consisting of -OR , -N(R ) 2 , -CN, -N0 2 , -X-R , -X-
  • Preferred compounds of formula I are tnose wherein each Y and Y' is independently selected from the
  • each R is independently selected from oxo, OR ,
  • each R is independently selected from the group consisting of R ; C ⁇ _-C 6 alkyl optionally substituted with R ; C 2 -C 6 alkenyl; C 2 -C 6 alkynyl; C 3 -C 6 cycloal ⁇ yI optionally fused with R ; C 5 -C 6 cycloalkenyl optionally fused with R 6; where any mem ⁇ bbe(r of R12 may be optionally ,2 substituted by one or more R
  • Preferred compounds of formula I are those wherein n is equal to 1; those having the structure of formula II:
  • Y is -(C(R ) 2 ) p -M-; those wherein X is -C(0)- or -S(0) 2 - and Y is (-C(R 2 ) 2 -) p ; those wherein X is -C(0)-, -C(0)C(0)- or -S(0) 2 -; and
  • Y is -N(R 2 )- or -N(R 2 )-CH 2 -.
  • An alternate object of this invention is a novel class of compounds represented by formula IV:
  • X and X' are independently -C(O)- or -S(0) 2 -;
  • Y is -(C(R 2 ) 2 )-M-, -(C(R 2 ) 2 ) p -r ⁇ N(R 2 )- or -N(R 2 )-
  • Another object of this invention is a novel class of compounds represented by formula V:
  • X is -C(O)- or -S (0) 2 -;
  • Y is -(C(R 2 ) 2 )-M-, -(C(R 2 ) 2 ) p -, -N(R 2 )- or -N(R 2 )-
  • R 10 is 0 or H 2 ; each R is independently H, OH or 0, wherein both
  • R ,11 are not simultaneously hydrogen;
  • Z is a structure of formula VI:
  • any structure of formula VI is optionally fused with an aryl, carbocyclic or heterocyclic ring and is optionally substituted with 1-3 substituents
  • R is H 2 , a methylene s implied); and each R 1 , R 2 , R 7 , R 4 , R 8 , p, q, G, M, Q and X' is independently as defined for formula I.
  • R 10 and R 11 are 0; compounds having the structure of formula V, wherein
  • R 10 and R 11 are 0; q is 1;
  • X' is -C(0)-; compounds having the structure of formula V, wherein
  • R 10 and R 11 are 0;
  • R is t-butylammo; compounds having the structure of formula V, wherein X is -C(0)-; (R 2 ) 2 ) p -; and compounds having the structure of formula V wherein X and X' is -C(O)-;
  • R is H 2 ; and one R is H and one R is OH;
  • X and X' is -C(O)-;
  • Y is -(C(R 2 ) 2 )-;
  • one R is H and one R is OH;
  • Y is -(C(R 2 ) 2 )-; R 7 is H; R 10 is H 2 ; one R is H and one R is OH; and
  • R 10 is H 2 ; one R is H and one R is OH; and
  • R within the definition of Y is selected from ;ne group consisting of:
  • R is H; one R is H and one R is OH; and
  • R within the definition of Y is aryl optionally substituted with one or more groups selected
  • X and X' is -C(O) -;
  • Y is -(C(R 2 ) 2 )-; R 7 is H;
  • R 10 is H 2 ; one R is H and one R s OH; and
  • Y is (C(R' R 10 is H 2 ;
  • R is C_-C_ alkyl optionally substituted with R ;
  • R 3 at least one R within the definition of Y is pyridyl, triazolyl, oxazolyl, isoxazolyl, pyrimidyl, pyrazolyl, pyridazmyl, thiazolyl, imidazolyl, thienyl thiadiazolyl, oxadiazolyl, triazmyl or pyrazmyJ
  • R may be optionally substituted with 1-3 substituents selected from -OR , -R , -N(R ) (R ) ,
  • R 10 is H 2 ; one R 1 1 is H and one R11 is OH;
  • R is benzyl optionally substituted with 1-3 substituents selected from -OR 9 , -N(R 9 ) (R 9 ), SR 9 , -X-R 9 ,
  • R is benzyl optionally substituted with 1-3 substituents selected from -OR 9 , -N(R 9 ) (R 9 ), SR 9 , -X-R 9 ,
  • each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected from the group consisting of oxo, -OR 9, -R9, -N(P9) (R9) ,
  • each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected
  • R within the definition of Y is selected from hydrogen, R or _-C ⁇ alkyl optionally substituted with
  • X and X' is -C(0)-
  • Y is -N(R 2 )-;
  • R is h 2 ; and one R is H and one R is OH; and those compounds of formula V wherein
  • X and X' is -C(O)-;
  • X is -C(O)- or -S(0) 2 -; and the compounds of formula IX wherein
  • X is -C(O)-
  • Y is - (C(R 2 ) 2 )-M-;
  • R is H; and those compounds of formula IX wherein
  • Y is -N(R 2 )-
  • R is H; and those compounds of formula IX wherein X is -C(O)-; Y is -(C(R 2 ) 2 A and R 7 is H.
  • X and X' are independently -C(O)- or -S(0) 2 -; those compounds of formula I having the structure of formula XII, wherein
  • X and X' are independently -C(O)- or -S(0) 2 ⁇ ; and 4 R is l-amino-2-hydroxyindanyl; and compounds of formula I having the structure of formula
  • X and X' are independently -C(0)- or -S(0) 2 -; compounds according formula I having the structure of formula XIII, wherein
  • X is -C(0)- or -S(0) 2 -;
  • X' is -C(0)-
  • Y is -(C(R 2 ) 2 ) ⁇ or -N(R 2 )-; and compounds of formula I having the structure of formula XIII, wherein
  • X is -C(0)-
  • Y is - (C(R 2 ) 2 )-; and R 7 is H; those compounds of formula XIII wherein X is -C(0)-; X' is -C(0)-;
  • Y is -(C(R 2 ) 2 )-; R 7 is H; and R within the definition of Y is selected from 3 hydrogen, R , or C ] _-Cg alkyl optionally substituted with
  • X' is -C(0)-
  • Y is -(C(R 2 ) 2 )-;
  • R is H
  • R within the definition of Y is selected from
  • X is -C(O)-
  • X' is -C(0)-
  • Y is - (C(R 2 ) 2 )-;
  • At least one R within the definition of Y is selected from the group consisting of:
  • R is H; and at least one R within the definition of Y is aryl optionally substituted with one or more groups selectee 9 c o from the group consisting of oxo, -OR , -R , -N(R" 1 (R " ), -N(R 9 )-X-R 9 , SR 9 , -X-R 9 , -0-X-N(R 9 ) 2 , -R 9 ⁇ OR 9 , -CN, -C0 2 R 9 , -X-N(R 9 ) (R 9 ) , halogen, -N0 2 , and -CF 3 ; those compounds according to formula XIII wherein:
  • X is -C(O)-
  • X' is -C(O)-
  • Y is -(C(R 2 ) 2 )-;
  • Y is -(C(R 2 ) 2 )-; at least one R within the definition of Y is py ⁇ dyl, triazolyl, oxazolyl, isoxazolyl, pyrimidyl, pyrazolyl, pyridazmyl, thiazolyl, imidazolyl, thienyl thiadiazolyl, oxadiazolyl, triaz yl or pyraz y wherein said R may be optionally substituted with 1-3 substituents selected from -OR 9, -R9, -N(R9) (R9),
  • X is -C(O)-
  • X' is -C(O)-
  • Y is -(C(R 2 ) 2 )-; R 7 is H; and R within the definition of Y is aryl optionally substituted with 1-3 substituents selected from -OR ,
  • each R not withm the definition of Y is 2- hydroxymdanyl.
  • each R not within the definition of Y is 2- hydroxymdanyl; those compounds accordmg to any of the aforementioned preferred compounds wherein: each RI is independently selected from benzyl optionally substituted with 1-3 substituents selected from the group consisting of OCH 3 , OH and NH 2 ; and those compounds accordmg to any of the aforementioned preferred compounds wherein: each RI is independently selected from benzyl optionally substituted with 1-3 substituents selected from the group consisting of 0CH 3 , OH and NH
  • each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected from the group consisting of oxo, -OR 9, -R9, -N(R9) (R9),
  • M, n, r, p, q and G is independently as defined for formula XIII .
  • Another embodiment is compounds accordmg to formula XIII, wherein:
  • R withm the definition of Y is selected from hydrogen, R or C ⁇ -Z 6 alkyl optionally substituted with R 3 ; each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected
  • each X, X', Y, Y', Z, R 1 , R 3 , R 4 , R 5 , R 7 , R 8 , R 9 , Q, M, n, r, p, q and G is independently as defined for formula XIII.
  • Another embodiment is compounds of formula I having the structure of formula XIII, wherein
  • Y is - (C (R 2 ) 2 )-; and compounds of formula I having the structure of fcrmula
  • Y is -N(R 2 )-; and R 7 is H.
  • preferred comDounds are those of formula V wherein
  • R 10 is H 2 ; and one R is H and one R is OH; and Z is selected from the group consisting of
  • R is as defined in formula I; and those of formula V wherein Z is selected from the group consisting of
  • R is H 2 ; and one R 11 is H and one R11 is OH.
  • R is H ; and one R11 is H and one R11 is OH, and those compounds of formula V, wherein
  • X and X' is -C (0)-;
  • R 7 is H
  • R is H ; and one R is H and one R s OH, and the aforementioned compounds of formula V wherein Z is selected from the group consisting of:
  • one R is H and one R is OH; and those compounds of formula V wherein X and X' is -C (0)-;
  • Y is -N(R 2 )-;
  • R 7 is H; one R is H and one R is OH, and those compounds of formula V, wherein X and X' is -C(0) -;
  • Y is -(C(R 2 ) 2 )-M-; M is 0; R 7 is H;
  • R is H 2 ; and one R 11 is H and one R11 is OH, and the aforementioned compounds of formula V wherein Z s selected from the group consisting of:
  • M, n, r, p, q and G is independently as defined for formula I.
  • R and R are defined as in formula I, comprising the steps of:
  • R is defined as in formula I, in an inert solvent, preferably an ethereal solvent such as diethyl ether or THF, with a base, preferably an alkali metal amide such as lithiumdiisopropylamide at a temperature between about -78 °C to about 25 °C;
  • an inert solvent preferably an ethereal solvent such as diethyl ether or THF
  • a base preferably an alkali metal amide such as lithiumdiisopropylamide at a temperature between about -78 °C to about 25 °C;
  • step (2) reacting the product of step (1) with an aldehyde R CHO followed by an optional treatment ' dehyrat g agent, preferably Martin's sulfurane dehydrating agent, wherein R is defined as m formula I to give a compound of formula XVI:
  • R and R are defined as in formula I;
  • step (3) reacting the product of step (2) m an inert solvent, preferably methanol, with hydrogen gas m the presence of an hydrogenation catalyst, preferably 10- palladium on carbon, followed by treatment with an anhydrous acid, preferably trifluoroacetic acid or 4N HCl in dioxane to give a product of formula XIV.
  • an inert solvent preferably methanol
  • hydrogenation catalyst preferably 10- palladium on carbon
  • R and R are as defined in formula I, in an inert solvent, preferably DMF or THF, with a base preferably sodium hydride, then bromomethylacrylic acid at a temperature between about -78 °C to about 25 °C; (2) reacting the product of step (1) with ar. oxidizing agent, preferably ozone and if necessary a reductive work-up with a reducing agent such as dimethylsulfide;
  • ar. oxidizing agent preferably ozone and if necessary a reductive work-up with a reducing agent such as dimethylsulfide
  • step (3) reacting the product of step (2) in an inert solvent, such as DMF, with thioproline t-butylamide and suitable amide-bond coupling reagents, preferably EDC,
  • R and r are defined as in formula I, comprising the steps of: 1) reacting a compound of formula XX
  • R ,1 i.s defined as in formula I and PG is a N- protecting group, such as those described in Greene and Wuts (infra) , preferably p-methoxybenzyl, an inert solvent, preferably THF, with a base, preferably lithiumdiisopropylamide at between about -78 °C to about 25 °C, then a bis-leaving group alkane of formula XXI:
  • LG is selected from halo, preferably chloro or o ⁇ o, arylsulfonate esters, preferably tosyl, and alkylsulfonate esters, preferably mesyl, and r is defined as in formula I, to give a product of formula XXII:
  • R and PG are defined as m formula XX and LG and r are defined as in formula XXI;
  • step (2) reacting the product of step (1) in an inert solvent, preferably THF, with a base, preferably lithiumdiisopropylamide, at between about -78 °C to about 25 °C to give a product of formula XXIII:
  • R is defined as in formula I and PG is a N- protecting group; (3) reacting the product of step (2) m an inert solvent with a reagent suitable for removal of the N- protecting group PG, such as those described in Greene and Wuts (inf,ra) . to give a compound of formula XIX.
  • R groups for formula I include:
  • Preferred compounds of this invention of formula I include the specific compounds contained i: Tables 1-5.
  • the preferred compounds of this invention are compound numbers (as in Tables 1-5) : 1, 2, 3, 4, 7, 8, 9, 13, 14, 16, 17, 18, 20, 23, 24, 25, 26, 32, 35, 38, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 62, 63, 72, 75, 76, 78, 80, 82, 83, 91, 92, 94, 95, 96, 101, 102, 109, 121, 122, 123, 124, 126, 127, 128, 129, 131, 132, 133, 134, 135, 137, 138, 140, 141, 145, 146, 147, 149, 150, 155, 156, 160, 161, 162, 164, 165, 170, 171, 175, 176, 177, 179, 180, 185, 186, 190, 191, 192, 194, 195, 200, 201, 208, 219, 220, 228 and 264.
  • compound numbers 2, 7, 8, 9, 14, 18, 20, 25, 26, 32, 38, 45, 47, 48, 49, 50, 51, 53, 54, 62, 63, 72, 82, 83, 91, 92, 94, 95, 96, 123, 126, 140, 141, 219, 220, 228 and 264. Even more preferred are compound numbers: 7, 8, 9, 20, 45, 50, 51, 53, 54, 82, 83, 92, 94, 96, 219, 220, 228 and 264.
  • this invention also relates to novel methods for preparing compounds and intermediates of the following structures.
  • One embodiment relates to a process
  • the compounds of this invention may be synthesized using conventional techniques. Advantageously, these compounds are conveniently synthesized from readily available starting materials. Although the syntheses of the compounds of this invention are known to those of skill in the art, the following general schemes are set forth to illustrate these methods. These schemes should not be viewed as limiting the scope of this invention m any way.
  • E2 is achieved by reaction of a starting aldehyde with ethyl diethylphosphoranylmethanesulfonate and subsequent reduction of the double bond (see: Gennari et al., Angew. Chem. Int. Ed. Engl., 33, pp. 2067-69 (1994)) to yield compound Ellla. Cyclization may then be achieved by deesterification and activation of the sulfonate moiety as described in Gennari, followed by deprotection of the nitrogen protection group to yield the cyclized product EIV. Alternatively, an ammo acid may be converted to compound Elllb using standard synthetic methods illustrated in Scheme 1. Compound Elllb can be cyclized to afford compound EIV. Compound EIV may then be N-protected, for example, the presence of Boc anhydride and DMAP (see: Flynn et al . , - I l l -
  • anion may then be quenched with a variety of electrophiles and subsequently deprotected to form the desired analogs of E2.
  • this anion may be quenched with an aldehyde to form (after subsequent dehydration, i.e., an aldol-type condensation) an exo-methylene compound which may then be reduced ⁇ i.e., hydrogenation) to form tne desired analogs of E2.
  • E3 results from a Wittig reaction using methyl (triphenylposphoranylidene) acetate followed by simultaneous reduction of the double bond and cyclization using magnesium metal methanol (Wei et al., Tetrahedron Lett., 34(28), pp. 4439-42 (1993)) .
  • a similar N-protection, deprotonation, quench and N- deprotection scheme, or condensation-reduction scheme, as described in the preparation of E2 results m desired analogs of E3.
  • E3 may be prepared from commercially available EVI.
  • the hydroxyl group may be activated using commonly available reagents such as methanesulfonyl chloride or para- toluenesulfonyl chloride m the presence of a tertiary amine base.
  • reagents such as methanesulfonyl chloride or para- toluenesulfonyl chloride m the presence of a tertiary amine base.
  • the addition of a nucleophile to displace the mesylate or tosylate yields EVII (Ackermann et al., Helv. Chim. Acta, 73, pp. 122-32 (1990)) which may be treated as described above to obtain E3.
  • activating reagents include but are not limited to carbonyldiimidazole, phosgene, sulf ryldichloride, sulfuryldiimidazole, sulfonyl dumide, and oxalyl chloride.
  • Methods leading to the production of analogs of compound E7 are also known m the art (McManus et al., J. Med. Chem., 8, pp. 766-76 (1965)) .
  • Scheme 3 exemplifies several potential routes to the synthesis of compound E7. Any protected ammo alcohol may be deprotonated to form the alkoxide which may be reacted with a substituted alpha bromo ester to form ether EIX
  • EIX may be formed from activation of a protected ammo alcohol with, for example, methanesulfonyl chloride or para- toluenesulfonyl chloride the presence on a tertiary amme base and subsequent addition of a nucleophile such as an alkoxide from an alpha hydroxy acid to displace mesylate or tosylate to yield EIX.
  • a nucleophile such as an alkoxide from an alpha hydroxy acid to displace mesylate or tosylate to yield EIX.
  • Compound EIX can then be deprotected, free based with a tertiary amme base or potassium carbonate in methanol, and heated to effect cyclization to form E7.
  • E7 may be prepared from a protected ammo alcohol by protection of the hydroxyl group with, for example, t-butyldimethyl silyl chloride/imidazole to afford the silyl ether. Subsequent nitrogen ⁇ eprotection and acylation with a alpha bromo acid in the presence of any number of available coupling agents (for example dicylcohexylcarbodiimide, other related carbodiimide reagents or isobutyl chloroformate) or acylation with an alpha bromo acid chloride provides compound EX.
  • coupling agents for example dicylcohexylcarbodiimide, other related carbodiimide reagents or isobutyl chloroformate
  • acylation with an alpha bromo acid chloride provides compound EX.
  • E7 may be prepared from the correspondmg a-methylene compound (i.e., both PJ are H in E7, the nitrogen may be protected if necessary) oy a multiple deprotonation-alkylation sequence to give an
  • each R may be attached to form a spirocyclic product (i.e., alkylation with a dihaloalkane) .
  • Schemes 4-6 describe methods for converting the cyclic compounds E1-E7 into compounds of this invention.
  • compounds of the type Z exemplified by compounds E1-E7
  • epoxides are readily synthesized via methods well known in the art (Maligres et al,, Tetrahedron Lett., 36, pp. 2195-98 (1995)) .
  • further modification of the compounds may be performed subsequent to epoxide opening using reactions and materials well known in the art. For example, subsequent to epoxide opening utilizing example EXIb deprotection of the carbamate allows further modification of the unmasked amme.
  • compounds EZ may be converted to the desired products m a more stepwise fashion.
  • Compounds EZ may be ⁇ eprotonated using, for example, sodium hydride in DMF and treated with a three carbon based epoxide to generate epoxide EXII.
  • reagents include, but are not limited to, eDibro ohy ⁇ rm, epichlorohydrin and glycidyl tosylate.
  • Scheme 6 describes a method for introduction of electrophiles into comounds of the type EXIV.
  • Said compounds may be protected with a variety of protecting groups, for example t-butyldimethylsilyl triflate, to mask the secondary hydroxyl group followed by treatment with a non-nucleophilic base such as lithium dusopropylamide or hexamethyldisilyzane to generate the anion alpha to the carbonyl.
  • Various electrophiles may then be added to substitute the position alpha to the carbonyl, or alternatively an aldol-type condensation-reduction scheme may be employed. Deprotection of the secondary hydroxyl then yields the desired ⁇ roduct.
  • the above synthetic schemes are not intended to comprise a comprehensive list of all means by which the compounds described and claimed in this application may be synthesized. Further methods will be evident to those of ordinary skill in the art.
  • Such modifications are known in the art and include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system) , increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
  • the compounds of this invention are characterized by a superior ability to inhibit protease activity and viral replication, particularly aspartyl protease activity. These compounds are especially well suited for inhibiting HIV aspartyl protease. We believe that this activity is due to specific steric and electronic interactions between the protease and compounds of this invention. This belief stems from our analysis of the structural basis for the activity of compounds of this invention, in view of the known crystal structures of HIV protease and bound inhibitors, such as the structure reported in Miller et al.
  • novel compounds of the present invention are excellent ligands for aspartyl proteases, particularly HIV-1 and HIV-2 proteases. Accordingly, these compounds are capable of targeting and inhibiting late stage events HIV replication, i.e., the processing of the viral polyprotems by HIV encoded proteases. Such compounds inhibit the proteolytic processing of viral polyprotem precursors by inhibiting aspartyl protease. Because aspartyl protease is essential for the production of mature virions, inhibition of that processing effectively blocks the spread of virus by inhibiting the production of infectious virions, particularly from chronically infected cells. Compounds accordmg to this invention advantageously inhibit the ability of the HIV-1 virus to infect immortalized human T cells over a period of days, as determined by an assay of extracellular p24 antigen — a specific marker of viral replication.
  • the compounds of this invention may be employed in a conventional manner for the treatment of viruses, such as HIV and HTLV, which depend on aspartyl proteases for obligatory events in their life cycle.
  • viruses such as HIV and HTLV
  • Such methods of treatment, their dosage levels and requirements may be selected by those of ordinary skill in the art from available methods and techniques.
  • a compound of this invention may be combined with a pharmaceutically acceptable ad uvant for administration to a virally-mfected patient in a pharmaceutically acceptable manner and m an amount effective to lessen the severity of the viral infection or to alleviate pathological effects associated with HIV infection or immunosuppression such as opportunistic infections or various cancers, tumors, CMV retmitis, Candida infections, maternal fetal transmission, and AIDS related dementia, .
  • the compounds of this invention may be used in prophylactics and methods for protecting individuals against viral infection durmg a specific event, such as childbirth, or over an extended period of time.
  • the compounds may be employed in such prophylactics either alone or together with other antiretroviral agents to enhance the efficacy of each agent.
  • the novel protease inhibitors of this invention can be administered as agents for treating or preventing HIV infection in a mammal.
  • the compounds of formula I may be readily absorbed into the bloodstream of mammals upon oral administration.
  • Compounds of formula I naving a molecular weight of less than about 600 g/mole and aqueous solubility of greater than or equal to 0.1 mg/mL are most likely to demonstrate high and consistent oral availability. This surprisingly impressive oral availability makes such compounds excellent agents for orally-administered treatment and prevention regimens against HIV infection.
  • the compounds of this invention may be administered to a healthy or HIV-infected patient either as a smgle agent or in combination with other anti-viral agents which interfere with the replication cycle of HIV.
  • a healthy or HIV-infected patient either as a smgle agent or in combination with other anti-viral agents which interfere with the replication cycle of HIV.
  • the compounds of this invention may be administered with other anti-viral agents which target different events in the viral life cycle and which target different viral substrams with varying susceptability to specific agents, the therapeutic effect of these compounds is potentiated.
  • the co-administered anti-viral agent can be one which targets early events in the life cycle of the virus, such as cell entry, reverse transcription and viral DNA integration into cellular DNA.
  • Anti-FIV agents targeting such early life cycle events lrclude, didanosme (ddl), dideoxycytidine (ddC) , d4T, zidovudme (AZT) , 3TC, 935U83, 1592U89, 524W91, polysulfated polysaccharides, sT4 (soluble CD4) , ganiclovir, trisodium phosphonoformate, eflornith e, ribavirm, acyclovir, alpha interferon and tri- methotrexate.
  • non-nucleoside inhibitors of reverse transcriptase may be used to potentiate the effect of the compounds of this invention, as may viral uncoatmg inhibitors, inhibitors of trans-activatmg proteins such as tat or rev, or inhibitors of the viral mtegrase.
  • Combination therapies accordmg to th s invention exert an additive or synergistic effect inhibiting HIV replication because each component agent of the combination acts on a different site of HIV replication or on different strains of virus present in an infectious population.
  • combination therapies may also advantageously reduce the dosage of a given conventional anti-retroviral agent which would be required for a desired therapeutic or prophylactic effect, as compared to when that agent is administered as a monotherapy.
  • Such combinations may reduce or eliminate the side effects of conventional smgle anti- retroviral agent therapies, while not interfering with the anti-retroviral activity of those agents. These combinations reduce potential of resistance to smgle agent therapies, while minimizing any associated toxicity.
  • HIV protease inhibitors may include viral population effects, whereby certain members of a virus population which show reduced sensitivity to one protease inhibitor may be fully sensitive to another inhibitor or may in fact have enhanced sensitivity to the second inhibitor.
  • administration of two or more different inhibitors may be used to reduce specific toxicities associated with a smgle agent.
  • This advantage of combination therapy also applies to co-admmistration of the protease inhibitor of this invention with other antiviral agents.
  • co-administration of more than one protease inhibitor may lower the rate of metabolic activation of the compounds of this invention, for instance, by inhibiting enzymatic systems such as cytochrome P 450 / or esterases or the like.
  • co-administration of compounds of this invention with protease inhibitors such as ritonavir or other agents such as ketoconazole, grapefruit juice and antiulcer medications such as H 2 -blockers, which inhibits cytochrome P4 50 3A 4 may advantageously enhance their biological half-life.
  • Combinatorial therapies include the administration of a compound of this invention with AZT, ddl, ddC, d4T,
  • the compounds of this invention may also be co-administered with other HIV protease inhibitors such as VX-478 (Vertex, also known as 141W94 (Glaxo-Wellco e) and KVX-478 (Kissei) ) , saqumavir (Ro 31-8959, Roche), indinavir (L-735,524, Merck) ), ritonavir ⁇ ABT 538, Abbott), nelfmavir (AG 1343, Agouron), palinavir (Bila 2011 BS) , U-103017 (Upjohn), XM 412 (DuPont Merck), XM 450 (DuPont Merck), BMS 186318 (Bristol-Meyers Squibb), CPG 53,437 (Ciba Geigy), CPG 61,755 (Ciba Geigy), CPG 70,726 (C oa Geigy), ABT 378 (Abbott),
  • retroviral reverse transcriptase inhibitors such as nucleoside derivatives, or other HIV aspartyl protease inhibitors, including multiple combinations comprising from 3-5 agents.
  • retroviral reverse transcriptase inhibitors such as nucleoside derivatives, or other HIV aspartyl protease inhibitors
  • the co-admmi tration of the compounds of this invention with retroviral reverse transcriptase inhibitors or HIV aspartyl protease inhibitors may exert a substantial additive or synergistic effect, thereby preventing, substantially reducing, or completely eliminating viral replication or infection or both, and symptoms associated tnerewith.
  • Particularly preferred is administration of a combination of a compound of formula I, 3TC and zidovud e (AZT) .
  • the compounds of this invention can also be administered in combination with immunomodulators and immunostimulators (e.g., bropirimme, anti-human alpha interferon antibody, IL-2, GM-CSF, interferon alpha, diethyldithiocarbamate, tumor necrosis factor, naltrexone, tuscarasol, and rEPO) ; and antibiotics (e.g., pentamid e lsethiorate) to prevent or combat infection and disease associated with HIV infections, such as AIDS, ARC and HIV-associated cancers.
  • immunomodulators and immunostimulators e.g., bropirimme, anti-human alpha interferon antibody, IL-2, GM-CSF, interferon alpha, diethyldithiocarbamate, tumor necrosis factor, naltrexone, tuscarasol, and rEPO
  • antibiotics e.g., pentamid e lsethiorate
  • the compounds of this invention When the compounds of this invention are administered in combination therapies with other agents, they may be administered sequentially or concurrently to the patient.
  • the additional agents may be administered separately, as part of a multiple dose regimen, from the compounds of this invention.
  • those agents may be part of a smgle dosage form, mixed together with the compounds of this invention in a single composition.
  • the pharmaceutical compositions accordmg to this invention may comprise a combination of an aspartyl protease inhibitor of this invention and one or more therapeutic or prophylactic agents.
  • the compounds of this invention can also be used as inhibitory agents for other viruses which depend on similar aspartyl proteases for obligatory events in their life cycle. These viruses include other AIDS-like diseases caused by retroviruses, such as simian immunodeficiency viruses, HTLV-I and HTLV-II.
  • the compounds of this invention may also be used to inhibit other aspartyl proteases, such as renin, pepsin, cymosin, RSV protease, AMV protease, SIV protease and FIV protease, and in particular, other human aspartyl proteases, including renin, and aspartyl proteases that process endothelin precursors.
  • aspartyl proteases such as renin, pepsin, cymosin, RSV protease, AMV protease, SIV protease and FIV protease
  • other human aspartyl proteases including renin, and aspartyl proteases that process endothelin precursors.
  • compositions of this invention comprise any of the compounds of the present invention, and pharmaceutically acceptable salts thereof, with any pharmaceutically acceptable carrier, adjuvant or vehicle.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d- ⁇ -tocopherol polyethyleneglycol 1000 succ ate, surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, polyethyleneglycol polymers such as PEG-400, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamme sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc
  • Cyclodextrins sucn as ⁇ -, ⁇ -, and ⁇ -cyclodextnn, or chemically modified derivatives such as hydroxyalkylcyclodextrms, including 2- and 3-hydroxypropyl- ⁇ -cyclodextrms, or other solublized derivatives may also be advantageously used to enhance delivery of compounds of formula I.
  • the pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vagmally or via an implanted reservoir. We prefer oral administration or administration by injection.
  • the pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes subcutaneous, mtracutaneous, intravenous, intramuscular, mtra- articular, intrasynovial, mtrasternal, mtrathecal, mtralesional, and mtracranial injection or infusion techniques.
  • the pharmaceutical compositions may be m the form of a sterile mjectable preparation, for example, as a sterile mjectable aqueous or oleaginous suspension.
  • This suspension may be formulated accordmg to techniques known m the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile mjectable preparation may also be a sterile mjectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example, as a solution in 1, 3-butaned ⁇ ol.
  • the acceptable vehicles and solvents that may be employed are mannitol, water. Ringer's solution and lsotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful m the preparation of mjectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as carboxymethyl cellulose or similar dispersing agents which are commonly used m the formulation of pharmaceutically acceptable dosage forms such as emulsions and or suspensions.
  • compositions of this invention may be orally administered in any ora] ly acceptable dosage form including, but not limited to, hard or soft gelatin capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and ⁇ ried corn starch.
  • the active ingredient may be suspended or dissolved in an oily phase combined with emulsifying and/or suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
  • compositions of this invention may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore w ll melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolve ⁇ in a carrier with suitable emulsifying agents.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved m a carrier.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or m a suitable enema formulation. Topically-transdermal patches are also mcluded m this invention.
  • compositions of thi s invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared accordmg to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizmg or dispersing agents known in the art.
  • Dosage levels of between about 0.01 ana about 100 mg/kg body weight per day, preferably between about 0.5 and about 75 mg/kg body weight per day of the active ingredient compound are useful in the prevention and treatment of viral infection, including HIV infection.
  • the pharmaceutical compositions of this invention will be administered from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5* to about 95% active compound (w/w) .
  • such preparations contain from about 20% to about 80 q active compound.
  • a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • the compounds of this invention are also useful as commercial reagents which effectively bind to aspartyl proteases, particularly HIV aspartyl protease.
  • the compounds of this invention, and their derivatives may be used to block proteolysis of a target peptide or may be derivatized to bind to a stable resin as a tethered substrate for affinity chromatography applications.
  • a compound of formula I may be tethered to an affinity column to purify recombinantly produced HIV protease.
  • Derivatization of the compounds of this invention to produce affinity chromatography res s and the methods used to purify proteases using such resms are well known and with the skill of the art.
  • TLC Thin layer chromatography
  • E. Merck silica gel 60 F25 plates and elution with the indicated solvent system. Detection of the compounds was carried out by treating the plate with an appropriate visualizing agent, such as 10% solution of phosphomolybdic acid in ethanol or a 0.1% solution of nmhydrin in ethanol, followed by heating, and/or by exposure to UV light or iodine vapors when appropriate.
  • Thick layer silica gel chromatography was also carried out using E. Merck 60 F 25 plates ("prep plates") of 0.5, 1.0, or 2.0 mm thickness.
  • Preparative HPLC was also carried out using C 18 reversed-phase media. HPLC retention times were recorded in minutes. NMR spectral data was recorded using a Bruker AMX500, equipped with either a reverse or QNP probe, at 500 MHz, and was taken in the indicated solvent.
  • test compounds were serially diluted in medium (RPMI 1640, 10% fetal calf serum (FCS) , and gentamycin) in 96-well plates (Costar 3598) using a Cetus Pro/Pette. Exponentially growing MT4 cells were harvested and centrifuged at 1000 rpm for 10 minutes in a Jouan centrifuge (model CR 4 12) . Cell pellets were resuspended in fresh medium (RPMI 1640, 20% FCS, 20% IL-2, and gentamycin) to a density of 5 x 105 cells/ml.
  • medium RPMI 1640, 10% fetal calf serum (FCS) , and gentamycin
  • Non ⁇ det-40 was added to each well of the incubation plate. After thorough mixing with a Costar multitip pipetter, 60 ul of the mixture was transferred to filter-bottomed 96-wellplates. The plates were analyzed in an automated assay instrument (Pandex Screen Machine, Baxter Biotechnology Systems) . The assay makes use of a propidium iodide dye to estimate the DNA content of each well.
  • the antiviral effect of a test compound is reported -as an IC 5 o, i.e. the inhibitory concentration that would produce a 501 decrease in the HIV induced cytopathic effect. This effect is measured by the amount of test compound required to restore 50% of the cell growth of HIV- mfected MT-4 cells compared to unmfected MT-4 cell controls.
  • syncytia formation a formation of a cell type and the desired readout, syncytia formation, reverse-transcriptase (RT) activity, or cytopathic effect as assayed by a dye uptake method may also be used as readouts of antiviral activity.
  • RT reverse-transcriptase
  • cytopathic effect as assayed by a dye uptake method may also be used as readouts of antiviral activity.
  • H. Mitsuya and S. Broder "Inhibition of the in vitro mfectivity and cytopathic effect of human T-lymphotropic virus type III/lymphoadenopathy- associated virus (HTLV-III/LAV) by 2', 3'- dideoxynucleosides", Proc. Natl. Acad. Sc . USA, vol. 83, pp. 1911-1915 (1986).
  • the compounds of this invention are able to inhibit the replication of the HIV virus m CD 4 cells of human lineage, they are of evident clinical utility for the treatment of HIV infection. These tests are predictive of the compounds ability to inhibit HIV protease __u. vivo.
  • the oxalyl chloride was added dropwise to a solution of DMSO in methylene chloride at -78 °C. After stirring for 10 minutes, the alcohol was added as a solution in methylene chloride. The reaction was then stirred at -78 °C for 45 minutes. At this time the triethylamine was added and a white precipitate formed. The reaction was then stirred 45 minutes at -78 °C and 45 minutes at 0 °C. The reaction was then quenched by the addition of a solution of 90g of citric acid in 300 mL of water. The organic portion of the reaction was then washed by (2 x 80 mL) of both saturated sodium bicarbonate and brine. The combined organic layers were then dried over sodium sulfate, filtered and concentrated in vacuo to leave a white solid. The aldehyde was then used without further purification the reductive amination.
  • Example 1A The aldehyde of Example 1A was dissolved in 180 mL of DMF at 25 °C. This was followed by addition of the aldehyde and 1.8 mL of acetic acid respectively. After 2 hours sodium cyanoborohydride was added, as a solid. The reaction was then stirred at 25 °C for 12 hours.
  • the reaction was then quenched by the addition ol 50 L of saturated sodium bicarbonate, and after 10 mm. diluted by 100 mL of diethyl ether. The organic portion was then washed by (2 x 50 mL) of both saturated sodium bicarbonate and brine. The combined organic layers were then dried over magnesium suLfate, filtered and concentrated vacuo.
  • the crude oil was purified by silica gel chromatography elutmg with 30 ethyl acetate: hexane to provide 8.8 g of product (2 mmol, 75%) .
  • Example IB The Boc amme of Example IB was stirred in 15 mL of 4N HCl at 25 °C for 1.5 hours. The reaction mixture was then concentrated in vacuo to provide a white foaming solid. 3.83 mg of the deprotected diamine was dissolved m 500 L of methylene chloride. To this, triethyl amme was added. After stirring for 20 minutes, CDI was added (solid) . The reaction was then stirred for 24 hours. This was followed by concentration vacuo. The crude material was purified by silica gel chromatography, eluting with ethyl acetate, to provide 2.15 g (67 %) of the desired allyl urea. E am le 2
  • Boc lactam 1 (1.27 g, leq) was dissolved in THF (27 mL) and cooled to -78 °C. To this solution was added LDA (Aldrich, 1.5 M in hexane, 3.7 mL, 1.2 eq) via syringe over 3 minutes. After stirring for 85 minutes at - 78 °C, a solution of ethyl iodoacetate (600 uL, 1.1 eq) in THF (13 mL) was added via syringe over 6 minutes. The reaction was then stirred at -78 °C for 4.5 hours, then at 1.5 hours at -40 C C.
  • LDA Aldrich, 1.5 M in hexane, 3.7 mL, 1.2 eq
  • the subsituted malonate 1 (1.62 mmol) was suspended m ethanol and to this was added cone. HCl (0.24 mL, 2.4 mmol) and 10% palladium on Carbon (0.162 mmol) . This mixture was then stirred under a balloon of hydrogen gas at room temperature overnight. The reaction was then filtered through Celite and to the filtrate was added triethylamine (10 L, excess) followed by solid sodium bicarbonate (excess) . The mixture was stirred for 0.5 hr, filtered and concentrated to yield a yellow solid. This residue was then dissolved m ethyl acetate and washed with water, 0.5N HCl, saturated sodium bicarbonate, and brine. The organic layer was dried (MgS0 4 ), filtered, and dried to yield crude lactam product 2, which was used as is.
  • Lactam 1 (1.18 mmol) was dissolved m ethanol (5mL) and to this solution was added KOH (10 mmol). The mixture was stirred for 3 hr at room temperature and then concentrated to dryness. The residue was dissolved m water and washed with diethyl ether. The aqueous layer was then acidified with HCl and extracted with ethyl acetate. The organic layer was dried (MgS04), filtered and concentrated in vacuo to yield 341 mg of a l ght yellow solid. The residue was dissolved in DMSO (3mL) and to this solution was added p-toluenesulfonic acid mono- hydrate, and the mixture was heated to 80 °C overnight.
  • the Cbz-phenylalaninol mesylate 1 (280 mg, 0.77 mmol) was stirred in acetonitrile (5 L) containing benzyl amine (413 mg, 3.85 mmol) and sodium iodide (115 mg, 0.77 mmol). The reaction was then refluxed for 24 hours. The reaction was then cooled to 25 °C and concentrated in vacuo. The crude oil was then purified by silica gel chromatography, eluting with CH 2 C1 2 with a gradient up to 1:1 CH 2 Cl 2 :EtOAc to provide 120 mg of the desired diamine 2.
  • the Cbz protected diamine 1 (120 mg, 0.32 mmol) was stirred in 2.0 mL of 30 % HBr in acetic acid for one hour. This was followed by concentration in vacuo, The crude oil was then dissolved into toluene and concentrated in vacuo two times followed by evacuation at approx. 1 mm Hg. The crude diamine was then purified by silica gel chromatography, eluting with 95:5:1, CH 2 C1 2 :MeOH:NH 4 OH to provide 71 g ( 90 %) of the desired diamine 2.
  • the reaction was then cooled to 25 °C and quenched by the addition of 3 mL of saturated sodium bicarbonate.
  • the reaction mixture was then diluted by 15 L of methylene chloride and washed by both saturated sodium bicarbonate and brine, (2 x 15 mL each) .
  • the organic portions were then dried over sodium sulfate, filtered and concentrated in vacuo .
  • the crude product was then purified by silica gel chromatography, eluting with 80% ethyl acetate : hexane to provide 35. 0 mg of the desired alcohol .
  • lactam 1.15g, 1.0 equiv. t-butyldimethylsilyl 1.5 equiv. + .5 eq. , trifluoromethanesulfonate (1.06mL) imidazole 2.5 equiv + .5 eq, (470mg!
  • Lactam 1 was dissolved 5mL of DMF and cooled to 0 °C. To this solution was then added imidazole followed by TBDMS-triflate. The reaction was then allowed to warm to room temperature. After approximatly 2 hours, an additional .5 eq. (80mg) of TBDMS-triflate and .5 eq. (265uL) of imidazole was added and the reaction was stirred overnight. The reaction was quenched with saturated NaHC0 3 solution and partitioned between H 2 0/EtOAc. The organic layer was washed with water(5X) and brme, ⁇ ried (MgS ⁇ ), filtered, and concentrated in vacuo to yield product 2 (1.5 gr,37%) which was used as is.
  • the reaction was partition betweem H 2 0/EtOAc and the organic layer was washed with with water and brine, drigd (MgS0 4 ) and filtered and concentrated in vacuo .
  • the residue was purified by silica gel chromatography (10% ether/ CH 2 C1 2) to yield benzyl product 2 (71mg, 48% ) .
  • Epoxide 1 was dissolved in 3 mL of EtOH and to this solution was added cylcopentylmethyla ine. The reaction was heated to 80 °C for 2.5 hours (TLC
  • Morpholinone 1 was dissolved in 1 ml of anhydrous DMF, cooled to 0 C and to this solution was added 4.4 mg of NaH. The solution was brought to room temperature for 30 min and then cooled down to OC before adding 0.20 g of epoxide 2. After heating for 5 hrs at 45 °C, the solvent was removed in vacuo and purified on silica gel yielding 111 mg of final product 2 (compound 47) .
  • M (ES+) 585 (M+l), 607.1 (M+Na) .
  • allyl lactam 1 0.80g was dissolved in 1 ml of DMF, cooled to 0 °C and 89.5 mg of sodium hydride was then added. The solution was then brought up to ambient temperature for 30 min, again cooled down to 0 °C and 1.4 g of epoxide 2 was added. The reaction was warmed to 50 °C under N 2 blanket for 3 hrs. The resulting crude mixture was then chromatographed on silica gel yielding 1.4g of 3 (63.7%). This amount was treated with 12 ml of 4N HCl in dioxane and 2 ml water for 30 min.
  • the desired material 3 was isolated as a minor fraction (9 mg) as a white solid 1H-NMR(CDC13) : 2.10(2H), 2.70(2H), 2.8-3.2(8H), 3.4(1H), 3.58(1H), 4.02(1H), 4.15(1H), 4.22(2H), 5.30(1H), 5.86(1H), 7.06(2H), 7.1-7.4 (16H) .
  • the oil was purified by column chromatography: hexane/ethyl acetate (60/40) to give 0.23g (50%) of the acetonide protected product.
  • the acetonide (0.185g, 0.29mmol) was dissolved in isopropanol (lOmL) and treated with cone. HCl (3.0mL) at room temperature. After 1.5h., the solution was adjusted to pH 11 with 3.ON NaOH and then concentrated. The aqueous solution was extracted with ethyl acetate (3 x75mL) . The ethyl acetate was dried (MgSO ⁇ ) and evaporatated to give a clear film.
  • Lactam epoxide 1 (180 mg, 0.66 mmol) and decahydroisoquinoline 2 (160 mg, 0.66 mmol) were heated to 80 °C in isopropanol. After three hours the reaction was cooled to 25 °C and stirred for 48 hours at room temperature. The reaction was then concentrated m vacuo . Purified by silica gel cnromatography, eluting with 25 % EtOAc : Hexanes, providing 90 mg (90% pure by HPLC) of desired product 3.
  • the reaction mixture was then diluted by the addition of 10 L of diethyl ether and washed by 2 x 10 mL of each saturated aqueous sodium bicarbonate and saturated aqueous brme.
  • the combined organic portions were then dried over MgS0 4 , filtered and concentrated in vacuo to provide the crude product.
  • Purification of the crude solid was carried out by silica gel chromatography (1000 uM S ⁇ 0 2 prep, plate) elut g with 20 % Me0H/CH 2 Cl 2 . This provided 3.1 mg of the desired product 2, with 96 % purity by HPLC.
  • the overall yield for addition, deprotection of N-Boc and coupling with 3-p ⁇ colyl chloride was 9 I .
  • the Boc protected piperazine 1 from the previous step was stirred for 1.5 hours in 1.0 L of 4 N HCl in dioxane. This was followed by concentration in vacuo.
  • the crude hydrochloride salt was then dissolved in 10 mL of CH 2 C1 2 and washed by 10 mL of both saturated sodium bicarbonate and saturated brine. The organic portion was then dried over MgS0 , filtered and concentrated in vacuo.
  • the free amine was then taken up in 1 mL of DMF. This was followed by the addition of 3-picolyl chloride HCl salt (50 mg, mmol) and triethyl amine (300 uL) , respectively. The reaction was then stirred at 25 °C for 30 hours.
  • the THF lactam 1 (0.4 mmol) was dissolved in dry DMF at 0 °C and to this solution was added sodium hydride (0.47 mmol) . After 30 min of stirring, (s)- epichlorohydrin (0.47 mmol) was added and the reaction was allowed to warm to room temperature and stir overnight. The reaction was then diluted with water and extracted with ethyl acetate. The organic layer was washed sequentially with 0.5N HCl, saturated NaHC0 3 and brine, followed by drying (MgS0 ), filtration and concentration in vacuo to yield product (118 mg, crude) which was used as is.
  • reaction mixture was diluted with 400 mL of dichloromethane, washe ⁇ once with 150 mL of water, dried (magnesium sulfate), filtered, concentrated, and purified on silica gel using 3:1 ethyl acetate/hexanes as the eluent, affording 0.6 g (46%) of the desired compound as a golden oil which solidified upon standing.
  • the lactam obtained above was coupled to the corresponding epoxide according to the protocol used for Example 24.
  • the final purification was performed on silica gel (2% 2M ammonia- ethanol in dichloromethane) to give the cis- and the trans- actam diastereomers each as white solids.
  • the iodolactam 1 (0.43 mmol) was dissolved in dry acetonitrile in a high pressure tube and to this solution was added diisopropylethylamme (Pierce, 0.65 mmol) followed by aniline 2 (Aldrich, 0.47 mmol) .
  • the tube was sealed and the reaction heated to 70 °C with stirring overnight.
  • the reaction was cooled to ambient temperature, solvent removed in vacuo, and the residue taken up in ethyl acetate/water.
  • the organic layer was washed sequentially with saturated aqueous NaHC03 and brme, followed by drying (MgS04), filtration and concentration in vacuo.
  • the PMB lactam 1 (0.46 mmol) was dissolved in dry THF at -78 °C and to this solution was added lithium diisopropylamide (Aldrich, 1.5 M in cyclohexane, 0.65 mmol). The solution was stirred for 15 minutes at -78 °C and 4- (Chloromethyl)-3, 5-d ⁇ methyl ⁇ soxazole 2 (Acros Organics, 0.56 mmol) was added. The cooling bath was removed and the solution warmed to room temperature and stirred overnight. The reaction was diluted with water and extracted with ethyl acetate.
  • Lactam 1 (0.13 mmol) was dissolved in 7:3 acetonitrile/water.
  • Ceric ammonium nitrate Aldrich, 0.26 mmol was added and the mixture was stirred at ambient temperature until the starting material was no longer evident by TLC.
  • Acetonitrile was removed in vacuo, and the residue taken up in ethyl acetate/water.
  • the organic layer was washed sequentially with saturated aqueous NaHC03 and brine, followed by drying (MgS04), filtration and concentration in vacuo .
  • Lactam 1 (1.43 mg, 4.86 mmol) was dissolved in anhydrous THF (25 mL) and cooled to -78 °C. This was followed by the addition of 3.9 mL of LDA (5.83 mmol,
  • the anion solution was stirred at -78 °C for 45 minutes and then cannulated into a -78 °C solution of p-formaldehyde (437 mg) in 25 mL of THF, washing with 1 mL of THF.
  • the reaction was warmed to room temperature over 4 hr and stirred overnight.
  • the reaction was quenched by the addition of 10 L of a saturated sodium bicarbonate, and concentrated in vacuo to remove the THF.
  • the crude reaction mixture was partitioned between ethyl acetate and saturated sodium bicarbonate. The aqueous layer was extracted with ethyl acetate.
  • the bromide (87.2 mg, 0.28 mmol) was dissolved in 2 mL of benzene and treated with imidazole (46mg, 3 EQ.). After heating to 125 °C for 20 hours the reaction was cooled to 25 °C and concentrated m vacuo .
  • the crude product which was purified by silica gel chromatography (5 % MeOH/CH 2 Cl 2 ) , to provide the addition product (50%) and the elimination product (2) in a 50 % yield.
  • the lactam 1 (621 mg, 2.02 mmol) was dissolved in 7 L acetonitrile, followed by the addition of H 2 0 (3 mL) . This was followed by the addition of CAN, 3.32 g (6.06 mmol, 3 EQ.) . The reaction was stirred at 25 °C for 1 hour. After concentrating the reaction in vacuo, the crude material was resuspended in ethyl acetate and washed with saturated sodium bicarbonate, brine, dried (MgS0 4 ) and filtered. Concentration in vacuo afforded the crude product which was purified by silica gel chromatography (3% methanol :CH 2 C1 2 ) to procide the desired unprotected lactam (122 mg, 32 %)
  • the , ⁇ -unsaturated lactam (55 mg, 0.29 mmol) was then heated to 130 °C in 2 ml of benzene containing imidazole (30 mg, 0.44 mmol) for 24 hours. After cooling to 25 °C, the reaction mixture was concentrate in vacuo . The crude material was purified by silica gel chromatography, eluting with 5% methanol :CH 2 C1 2 to provide 46.7 mg of the desired addition product (63 %) as well as 15.7 mg of recovered starting olefin (29 %) .
  • the iodolactam 1 (0.45 mmol) was dissolved in dry acetonitrile in a high pressure tube and to this solution was added diisopropylethylamme (Pierce, 1.35 mmol) followed by indoline 2 (Aldrich, 0.54 mmol) .
  • the tube was sealed and the reaction heated to 70 °C with stirring overnight.
  • the reaction was cooled to ambient temperature, solvent removed m vacuo, and the residue taken up in ethyl acetate/water.
  • the organic layer was washed sequentially with saturated aqueous NaHC03 and brme, followed by drying (MgS04), filtration and concentration m vacuo .
  • Triethylamine (2.0 mL, 14.3 mmol) was added dropwise. The solution was refluxed for 15 minutes and cooled. A white solid precipitated durmg the heating period.
  • isoxazole PMB lactam 1 (900 g, 2.30 mmol) was dissolved in 14 mL of 70% CH 3 CN-H 2 0. Ceric ammonium nitrate (3.607 g, 6.58 mmol) was added forming a dark orange solution. The mixture was stirred until the starting material was no longer evident by TLC (10% EtOAc/CH 2 Cl 2 ) . The light yellow solution was diluted with CH 2 C1 2 and washed with water.
  • the iodolactam 1 (0.78 mmol) was dissolved in dry acetonitrile m a high pressure tube and to this solution was added diisopropylethylamme (Pierce, 2.35 mmol) followed by N-methylanilme 2 (Aldrich, 0.94 mmol) .
  • the tube was sealed and the reaction heated to 70 °C with stirring overnight.
  • the reaction was cooled to ambient temperature, solvent removed m vacuo, and the residue taken up m ethyl acetate/water.
  • the organic layer was washed sequentially with saturated aqueous NaHC03 and brme, followed by drying (MgS04), filtration and concentration m vacuo .
  • the PMB- lactam 1 (563.7 mg, 2.75 mmol) was dissolved m 10 mL of THF.
  • the solution was cooled to -78 °C and 1.5M LDA (2.0 mL, 3.00 mmol) was added dropwise via syringe producing the yellow color of the enolate.
  • the solution was stirred for 15 minutes at -78 °C and propargyl bromide (310 uL, 3.48 mmol) was added dissipating the yellow color.
  • the cooling bath was removed and the solution was warmed to room temperature and stirred overnight.
  • the solution was poured into IN HCl and extracted with CH 2 C1 2 .
  • the iodolactam 1 (1.38 mmol) was dissolved in dry acetonitrile in a high pressure tube and to this solution was added diisopropylethylamine (Pierce, 4.15 mmol) followed by tetrahydroquinoline 2 (Aldrich, 1.66 mmol) .
  • the tube was sealed and the reaction heated to 70 °C with stirring overnight.
  • the reaction was cooled to ambient temperature, solvent removed in vacuo, and the residue taken up .in ethyl acetate/water.
  • the organic layer was washed sequentially with saturated aqueous NaHC03 and brine, followed by drying (MgS04) , filtration and concentration in vacuo .
  • N- chlorosuccinimide (2.5177 g, 18.9 mmol) was dissolved in 75 mL of CH 2 C1 2 .
  • the solution was cooled to 0 C C and thiophenol (1.90 mL, 18.5 mmol) was added dropwise via syringe causing an immediate formation of a yellow color and an exotherm.
  • the orange solution of PhSCI was stirred for 30 minutes at room temperature and a solution of the allyl lactam 1 (6.156 g, 18.4 mmoP was added dropwise dissipating the orange color.
  • the light yellow solution was stirred for two hours and the solvent was removed in vacuo.
  • trimethylsilyl diazomethane 140 uL, 0.280 mmol was dissolved in 5 mL of THF.
  • the bright yellow solution was cooled to -78 C C and n-BuLi (320 uL, 480 mmol) was added.
  • the vinyl sulfone PMB lactam 1 108 mg, 0.227 mmol was dissolved in 5 mL of THF and added dropwise via syringe at -78 °C to the lithiate solution.
  • the TMS pyrazole PMB lactam 1 (1.1345 g, 2.53 mmol) was dissolved in 110 mL of 91% CH 3 CN/H 2 0.
  • Tetrabutylammonium fluoride (2.7 mL of a 1.0M solution in THF, 2.70 mmol) was added dropwise via syringe. The reaction was refluxed for 48 hours and cooled. The solvent was evaporated in vacuo and the residue was dissolved in CH 2 C1 2 .
  • the pyrazole PMB lactam 1 (588 mg, 1.57 mmol) was dissolved in 25 mL of THF. NaH (50 mg of a 60% dispersion in mineral oil, 2.08 mmol) was added. Gas evolution was observed. Methyl chloroformate (140 uL, 1.81 mmol) was added and the reaction was stirred at room temperature overnight. The mixture was acidified with IN HCl and extracted with CH 2 C1 . The organic extract was dried (MgS0 4 ), and evaporated in vacuo to afford the pyrazole carbamate PMB lactam 2 (588 mg, 87%) as a light yellow oil .
  • the pyrazole carbamate PMB lactam 1 (577 mg, 1.33 mmol) was dissolved in 30 mL of 70% CH 3 CN-H 2 0. Ceric ammonium nitrate (2.5123 g, 4.58 mmol) was added. The orange solution was stirred at room temperature until the starting material was no longer evident by TLC (1 hr) . The light yellow solution was poured into water and extracted with EtOAc. The organic extract was dried (MgS0 4 ) and evaporated vacuo to afford the pyrazole carbamate lactam 2 (228 mg, 55%) as a clear, colorless oil.
  • the benzyl triazole PMB lactam 1 (740 mg, 1.59 mmol) was dissolved 22 mL of 70% CH 3 CN-H 2 0. Ceric ammonium nitrate (2.1 g, 3.83 mmol) was added. The orange solution was stirred at room temperature until the starting material was no longer evident by TLC (1 hr) . The mixture was poured into water and extracted with EtOAc. The organic extract was dried (MgS0 4 ) and evaporated vacuo to afford the benzyl triazole lactam 2 (336 mg, 61%) as a clear, colorless oil.
  • BOC-lactam 1 (1.8 g, 6.6 mmol) was dissolved in THF (50 mL) and cooled to -78 °C. To this solution was added LDA (Aldrich, 1.5 M in cyclohexane, 5.3 mL, 7.9 mmol) via syringe over 10 minutes. After stirring for 60 mm at -78 C C, acetone (4.9 mL, 66 mmol) was added via syringe over 1 minute. The reaction was stirred for an additional 40 minutes before being quenched with IN HCl (15 mL) . Ethyl acetate (100 mL) was added and the layers were partitioned.
  • LDA Aldrich, 1.5 M in cyclohexane, 5.3 mL, 7.9 mmol
  • the alkene, 10% Pd-C (1.0 g) , and methanol (40 mL) were combined in a Parr bottle and pressurized to 50 psi of hydrogen gas. After 4 h of agitation, the reaction vessel was evacuated and filtered through a plug of Celite. The cake was washed with ethyl acetate (20 mL) and the combined filtrate was concentrated in vacuo to give the isopropyl BOC-lactam as a pale yellow oil. The lactam was dissolved in dichloromethane (20 mL) and trifluoroacetic acid (10 mL) was added slowly.
  • reaction mixture was diluted with 400 L of dichloromethane, washed IX with 150 mL of water, dried (magnesium sulfate) , filtered, concentrated, and purified on silica gel using 3:1 ethyl acetate/hexanes as the eluent affording 0.6 g (46%) of the desired compound as a golden oil which solidified upon standing.
  • the first step of the sequence was performed as for Example 43.
  • the olefin was carried forward as follows:
  • step 2 A vigorously stirred suspension of 330 mg (1.25 mmoL) of eneamide and 80 mg of 10% palladium on carbon (Degussa) in 12mL of anhydrous methanol was hydrogenated (Hydrogen balloon) for 1 h. The mixture was diluted with 100 mL of methanol, carefully filtered, concentrated, and purified on silica gel using ethyl acetate as the eluent affording 295 mg (89%) of an isomeric mixture of the desired compounds as a golden oil which solidified upon standing.
  • the 4-pyridylmethylpyrrolidone was prepared following procedures outline for Example 44.
  • the reaction was quenched with 10% aqueous solution of citric acid and extracted to ethyl acetate, and the product treated with 1:1 (v/v) DCM/TFA (40 mL) for 40 mm, after which solvents were removed and the crude purified to homogeneity by RP HPLC with total yield of 14.2%.
  • the resulting TFA salt was then neutralized with triethylamine, extracted between ethyl acetate/water, organics collected and dried, thus yielding a free base form of the spiropyrrolidone product which is used in subsequent coupling to the epoxide.

Abstract

This invention relates to a novel class of compounds of formula (I) that are aspartyl protease inhibitors. In one embodiment, this invention relates to a novel class of aspartyl protease inhibitors characterized by specific structural and physicochemical features. This invention also relates to pharmaceutical compositions comprising these compounds. The compounds and pharmaceutical compositions of this invention are particularly well suited for inhibiting HIV-1 and HIV-2 protease activity and consequently, may be advantageously used as anti-viral agents against the HIV-1 and HIV-2 viruses. This invention also relates to methods for inhibiting aspartyl protease activity and methods for treating viral infections using the compounds and compositions of this invention. A compound according to formula (I) wherein each Z is (a) or (b) or (c) wherein any Z may be optionally fused with R6; each X and X' is independently selected from the group consisting of C-C(O)-, -C(O)C(O)-, -S(O)- and -S(O)¿2?; each Y and Y' is independently selected from the group consisting of -(C(R?2)¿2)p-, -NR2-, -(C(CR2)2)p-M-, C=C(R2)2, and -N(R2)-CH2-.

Description

ASPARTYL PROTEASE INHIBITORS
TECHNICAL FIELD OF THE INVENTION The present invention relates to a novel class of compounds which are aspartyl protease inhibitors. In one embodiment, this invention relates to a novel class of HIV aspartyl protease inhibitors cnaractenzed by specific structural and physicochemical features. This invention also relates to pharmaceutical compositions comprising these compounds. The compounds and pharmaceutical compositions of this invention are particularly well suited for inhibiting HIV-1 and HIV-2 protease activity ana consequently, may be advantageously used as anti- viral agents against the HIV-1 and HIV-2 viruses. Tms invention also relates to methods for inhibiting aspartyl protease activity, methods for treating viral infections using the compounds and compositions of this invention, and methods for making intermediates and compounds of this invention. BACKGROUND OF THE INVENTION The human immunodeficiency virus ("HIV") is the causative agent for acquired immunodeficiency syndrome ("AIDS") — a disease characterized by the destruction of the immune system, particularly of CD4
T-cells, with attendant susceptibility to opportunistic infections — and its precursor AIDS-related complex ("ARC") — a syndrome characterized by symptoms such as persistent generalized lymphadenopathy, fever and weight loss.
As m the case of several other retroviruses, HIV encodes the production of a protease which carries out post-translational cleavage of precursor polypeptides in a process necessary for the formation of infectious viπons (S. Crawford et al., "A Deletion Mutation in the 5' Part of the pol Gene of Moloney Murine Leukemia Virus Blocks Proteolytic Processing of the gag and pol Polyprotems", J. Virol., 53, p. 899 (1985) ) . These gene products include pol. which encodes the virion RNA-dependent DNA polymerase
(reverse transcriptase) , an endonuclease, HIV protease, and gag, which encodes the core-proteins of the virion (H. Toh et al., "Close Structural Resemblance Between Putative Polymerase of a Drosophila Transposable Genetic Element 17.6 and pol gene product of Moloney Murine Leukemia Virus", EMBO J. , 4, p. 1267 (1985); L.H. Pearl et al. , "A Structural Model for the Retroviral Proteases", Nature, pp. 329-351 (1987); M.D. Power et al., "Nucleotide Sequence of SRV-1, a Type D Simian Acquired Immune Deficiency Syndrome Retrovirus",
Figure imgf000004_0001
A number of synthetic anti-viral agents have been designed to target various stages in the replication cycle of HIV. These agents include compounds which block viral binding to CD4 T- lymphocytes (for example, soluble CD4) , and compounds which interfere with viral replication by inhibiting viral reverse transcriptase (for example, didanosine and zidovudme (AZT) ) and inhibit integration of viral DNA into cellular DNA (M.S. Hirsh and R.T. D'Aqulia, "Therapy for Human Immunodeficiency Virus Infection", N.F.nσ.J.Med. , 328, p. 1686 (1993)). However, such agents, which are directed primarily to early stages of viral replication, do not prevent the production of infectious virions in chronically infected cells. Furthermore, administration of some of these agents m effective amounts has led to cell-toxicity and unwanted side effects, such as anemia and bone marrow suppression.
More recently, drug design efforts have been directed toward creating compounds which inhibit the formation of infectious virions by interfering with the processing of viral polyprotem precursors. Processing of these precursor proteins requires the action of virus-encoded proteases which are essential for replication (Kohl, N.E. et al. "Active HIV Protease is Required for Viral Infectivity" Proc. Natl. Acad. Sci. _____, 85, p. 4686 (1988)). The anti-viral potential of HIV protease inhibition has been demonstrated using peptidal inhibitors. Such peptidal compounds, however, are typically large and complex molecules that tend to exhibit poor bioavailability and are not generally consistent with oral administration. Accordingly, the need still exists for compounds that can effectively inhibit the action of viral proteases, for use as agents for preventing and treating chronic and acute viral infections. Such agents would be expected to act as effective therapeutic agents m their own right. In addition, since they act at a separate stage in the virus life cycle from previously described antiretroviral agents, the administration of a combination of agents would be expected to result m increased therapeutic efficacy.
SUMMARY OF THE TNVF.NTTON The present invention provides a novel class of compounds, and pharmaceutically acceptable derivatives thereof, that are useful as inhibitors of aspartyl proteases, and m particular, HIV aspartyl protease. The compounds of this invention can be used alone or in combination with other therapeutic or prophylactic agents, such as anti-virals, antibiotics, immunomodulators or vaccines, for the treatment or prophylaxis of viral infection.
According to a preferred embodiment, the compounds of this invention are capable of inhibiting HIV viral replication in human CD cells including T- cells, monocytic lines including macrophages and dendrocytes and other permissive cells. These compounds are useful as therapeutic and prophylactic agents to treat or prevent infection by HIV-1 and related viruses which may result in asymptomatic infection, AIDS-related complex ("ARC"), acquired immunodeficiency syndrome ("AIDS"), or similar d sease of the immune system.
It is a principal object of this invention to provide a novel class of compounds that are aspartyl protease inhibitors, and particularly, HIV aspartyl protease inhibitors. This novel class of compounds is represented by formula I:
Figure imgf000007_0002
wherein
Figure imgf000007_0001
wherein any Z may be optionally fused with R ; each X and X' is independently selected from the group consisting of -C (O) - , -C (0) C (0) - , -S (0) - and
-s {0) 2 ; each Y and Y' is independently selected from the group consisting of -(C(R )2)p-, -NR -/ - (C (R2) 2)p~M-, >C=C(R2)2, and -N(R2)-CH2-; each R is independently selected from the group consisting of hydrogen; R ; C1-C6 alkyl; C2-C6 alkenyl; C2-C6 alkynyl; C3-C6 cycloalkyl optionally fused with R ; C5-C6 cycloalkenyl optionally fused with R ; and where R 's are attached to adjacent atoms, the Rlfs together with their attached adjacent atoms form a carbocyclic or heterocyclic ring system which may be optionally fused with R ; where any member of R may be optionally substituted by one or more R ; 2 each R is independently selected from hydrogen;
R ; C_-C6 alkyl; C2-C6 alkenyl; C2-C6 alkynyl; C3-C6 cycloalkyl optionally fused with R ; C5-C6 cycloalkenyl optionally fused with R ; and where two R 's are
2 attached to the same gemmal atom, the R 's together with their attached geminal atom may form a spirocarbocyclic or spiroheterocyclic ring system;
2 where any member of R may be optionally substituted by
3 one or more R ; each R 3 is independently selected from oxo, OR9,
N(R9)2, N(R9)-X-R9,N(R9)-X-OR9, N(R9) -X-N (R9) 2, SR9, X-
R9, 0-X-N(R9)2r C(0)N(R9)2, halogen, N02, CN, COOR9 and
R6;
4 each R is independently selected from from the group consisting of OR9; N(R9)2; X-R9; C(0)N(R9)2; R6;
A~C6 alkyl; C2-C4 alkenyl; C3-C6 cycloalkyl optionally fused with R ; 5 -C Q cycloalkenyl optionally fused with
6 4
R ; where any member of R may be optionally substituted by one or more groups independently
9 3 selected from the group consisting of R and R ;
5 each R is independently selected from the group consisting of H, OH, 0 and R ; each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl may be optionally substituted with one or more groups selected
9 9 9 9 from the group consisting of oxo, -OR , -R , -N(R ) (R ),
-N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-0R9, -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3; 7 each R is independently selected from the group consisting of hydrogen, OH and 0; p each R is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, and heterocyclyl; each R is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heterocyclyl, aralkyl, carbocyclylalkyl and heterocyclylalkyl wherein any aryl, carbocyclyl or p heterocyclyl may be optionally fused with R and
Q wherein any member of R may be optionally substituted by one or more groups independently selected from the
8 8 fi group consisting of -OR , -N(R ) 2 , -CN, -N02, -X-R , -X- N(R8)2, -C(0)0R8, -N(R8)-XNR8, and halogen; each Q is independently selected from CH and N; each M is independently selected from the group
2 consisting of NH, -NR -, -0-, -S-, -S (O) - and -S(0)2~;
Figure imgf000009_0001
each r is 0,1 or 2; each p is independently 1 or 2; each q is independently 1, 2 or 3; and each G is independently selected from the group
2 consisting of -NH-, -NR -, -0-, -S-, -S(O)-, S(0)2,
-C(0)-, and -C(R2)2~. An alternate object of this invention is a novel class of compounds represented by formula IV:
Figure imgf000009_0002
wherein:
X and X' are independently -C(O)- or -S(0)?-,
Y is -(C(R2)2)-M-, -(C(R2)2; -N(R2)- or -N(R2)
CH and each R1, R2, R7, R4, p and M is independently as defined for formula I.
Another object of this invention is a novel class of compounds represented by formula V:
Figure imgf000010_0001
wherein:
X is -C(O)- or -S(0)2-;
Y is -(C(R2)2)-M-, -(C(R2)2)p-, -N(R2)- or -N(R2)-
CH2-;
R10 is 0 or H2; each R is independently H, OH or 0, wherein both
R ,11 are not simultaneously hydrogen; Z is a structure of formula VI:
Figure imgf000010_0002
(VI)
wherein any structure of formula VI is optionally fused with an aryl, carbocyclic or heterocyclic ring and is optionally substituted with 1-3 substituents independently selected from R ; and each R 1, R2
Figure imgf000010_0003
R8, G, M, Q and X' is independently as defined for formula I - sa ¬
lt is also an object of this invention to provide pharmaceutical compositions comprising the compounds of formulas I, IV and V and methods for their use as inhibitors of aspartyl protease, and particularly, HIV aspartyl protease.
It is a further object of this invention to provide methods for treating viral diseases, and in particular HIV-related diseases, using the compounds and compositions of this invention.
DETAILED DESCRIPTION OF THE INVENTION In order that the invention herein described may be more fully understood, the following detailed description is set forth. In the description, the following abbreviations are used:
Designa ion Reaσent or Fragment
Ac acetyl
Me methyl
Et ethyl
Bn benzyl
Trityl triphenylmethyl
Asn D- or L-asparagme
He D- or L-isoleucme
Phe D- or L-phenylalamne
Val D- or L-valme
Boc tert-butoxycarbonyl
Cbz benzyloxycarbonyl (carbobenzyloxy)
Fmoc 9-fluorenylmethoxycarbonyl
DCC dicyclohexylcarbodiimide
DIC diisopropylcarbodiimide
EDC 1- (3-dιmethylammopropyl) -3- ethylcarbodnmide hydrochloride
HOBt 1-hydroxybenzotrιazole
HOSu 1-hydroxysuccιnιmιde
TFA trifluoroacetic acid
DIEA dusopropylethy1amine
DBU l,8-dιazabιcyclo(5.4.0) undec-7-ene
EtOAc ethyl acetate t-Bu tert-butyl lBu iso-butyl
DMF dimethylformamide
THP tertrahydropyran
THF tetrahydrofuran
DMSO dimethylsulfoxide The following terms are employed herein:
Unless expressly stated to the contrary, the terms "-S0 -" and "-S(0)2-" as used herein refer to a sulfone or sulfone derivative (i.e., both appended groups linked to the S), and not a sulfinate ester.
The term "alkoxy" refers to an alkyl ether radical, wherein the term "alkyl" is as defined above. Examples of suitable alkyl ether radicals include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy, tert- butoxy and the like.
The term "alkyl", alone or in combination with any other term, refers to a straight-chain or branch-chain saturated aliphatic hydrocarbon radical containing the specified number of carbon atoms, or where no number is specified, preferably from 1-10 and more preferably from 1-5 carbon atoms. Examples of alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec- butyl, tert-butyl, pentyl, isoamyl, n-hexyl and the like.
The term "alkenyl", alone or in combination with any other term, refers to a straight-chain or branched-chain mono- or poly-unsaturated aliphatic hydrocarbon radical containing the specified number of carbon atoms, or where no number is specified, preferably from 2-10 carbon atoms and more preferably, from 2-6 carbon atoms. Examples of alkenyl radicals include, but are not limited to, ethenyl, E- and Z-propenyl, isopropenyl, E- and Z-butenyl, E- and Z-isobutenyl, E- and Z-pentenyl, E- and Z-hexenyl, E,E-, E,Z-, Z,E- and Z,Z-hexadienyl and the like.
The term "anti-viral agent" or "anti- retroviral agent" refers to a compound or drug which possesses viral inhibitory activity. Such agents include reverse transcriptase inhibitors (including nucleoside and non-nucleoside analogs) and protease inhibitors. Preferably the protease inhibitor is an HIV protease inhibitor. Examples of nucleoside analog reverse transcriptase inhibitors include, but are not limited to, zidovudme (AZT) , dideoxycytidme (ddC) , didanosme (ddl), stavudme (d4T) , 3TC, 935U83, 1592U89 and 524W91. Examples of non-nucleoside analog reverse transcriptase inhibitor include, but are not limited to TIBO, delavirdme (U90) and nevirapme. Examples of HIV protease inhibitors include, but are not limited to VX-478 (Vertex, also known as 141 94 (Glaxo-Wellcome) and KVX-478 (Kissei)), saqumavir (Ro 31-8959, Roche), mdmavir (L-735,524, Merck)), πtonavir (ABT 538,
Abbott), nelfmavir (AG 1343, Agouron), palinavir (Bila 2011 BS) , U-103017 (Upjohn), XM 412 (DuPont Merck), XM 450 (DuPont Merck), BMS 186318 (Bristol-Meyers Squibb), CPG 53,437 (Ciba Geigy) , CPG 61,755 (Ciba Geigy) , CPG 70,726 (C ba Geigy), ABT 378 (Abbott), GS 3333 (Gilead Sciences) , GS 3403 (Gilead Sciences), GS 4023 (Gilead Sciences), GS 4035 (Gilead Sciences), GS 4145 (Gilead Sciences), GS 4234 (Gilead Sciences), and GS 4263 (Gilead Sciences) . The term "aryl", alone or in combination with any other term, refers to a carbocyclic aromatic radical (such as phenyl or naphthyl) containing the specified number of carbon atoms, preferably from 6-14 carbon atoms, and more preferably from 6-10 carbor atoms. Examples of aryl radicals include, but are not limited to phenyl, naphthyl, indenyl, mdanyl, azulenyl, fluorenyl, anthracenyl and the like.
The term "carbocycle" and "carbocyclyl" radical, refers to a non-aromatic stable 3- to 8- membered carbon ring which may be saturateα, ono- unsaturated or poly-unsaturated. The carbocycle may be attached at any endocyclic carbon atom which results in a stable structure. Preferred carbocycles have 5-6 carbons.
The term "heterocycle" and "heterocyclyl" radical, unless otherwise defined herein, refers to a stable 3-7 membered monocyclic heterocyclic ring or 8- 11 membered bicyclic heterocyclic ring which is either saturated or unsaturated, and which may be optionally benzofused if monocyclic. Each heterocycle consists of one or more carbon atoms and from one to four heteroatoms selected from the group consisting of nitrogen, oxygen and sulfur. As used herein, the terms "nitrogen and sulfur heteroatoms" include any oxidized form of nitrogen and sulfur, and the quaternized form of any basic nitrogen. In addition, any ring nitrogen may be optionally substituted with a substituent R , as defined herein for compounds of formula I . A heterocyclyl radical may be attached at any endocyclic carbon or heteroatom which results in the creation of a stable structure. Preferred heterocycles include 5-7 membered monocyclic heterocycles and 8-10 memebered bicyclic heterocycles. Preferred heterocycles defined above include, for example, benzimidazolyl, imidazolyl, lmidazolinoyl, lmidazolidinyl, quinolyl, isoquinolyl, indolyl, mdazolyl, mdazolmolyl, perhydropyridazyl, pyridazyl, pyridyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazmyl, qumoxolyl, piperidmyl, pyranyl, pyrazolmyl, piperazmyl, pyrimidmyl, pyridazmyl, morpholinyl, thiamorpholmyl, furyl, thienyl, triazolyl, thiazolyl, β-carbolinyl, tetrazolyl, thiazo- lidmyl, benzofuranoyl, thiamorphol yl sulfone, oxazolyl, benzoxazolyl, oxopiperid yl, oxopyrrold yl, oxoazepinyl, azepinyl, isoxazolyl, isothiazolyl, furazanyl, tetrahydropyranyl, tetrahydrofuranyl, thiazolyl, thiadiazoyl, dioxolyl, diox yl, oxath:olyl, benzodioxolyl, dithiolyl, thiophenyl, tetrahydrothiophenyl and sulfolanyl, dioxanyl, dioxolanyl, tetrahydrof rodihydrofuranyl, tetrahydropyranodihydrofuranyl, dihydropyranyl, tetrahydrofurofuranyl and tetrahydropyranofuranyl .
The term "halogen" refers to a radical of fluorine, chlorine, bromine or iodine.
The terms "HIV protease" and "HIV aspartyl protease" are used interchangeably and refer to the aspartyl protease encoded by the human immunodeficiency virus type 1 or 2. In a preferred embodiment of this invention, these terms refer to the human immunodeficiency virus type 1 aspartyl protease.
The term "inert solvent" refers to a solvent reaction medium which allows the reagents to react together at a substantially increased rate relative to any reagent reacting with the designated solvent.
The term "leaving group" or "LG" refers to groups readily displaceable by a nucleophile, such as an amine, alcohol, phosphorous or thiol nucleophile or their respective anions . Such leaving groups are well known and include carboxylates, N-hydroxysuccinimide, N-hydroxybenzotriazole, halogen (halides) , triflates, tosylates, mesylates, alkoxy, thioalkoxy, phosph ates, phosphonates and the like. Other potential nucleophiles include organometallic reagents known to those skilled in the art.
The term "protecting group" refers to a suitable chemical group which may be attached to a functional group and removed at a later stage to reveal the intact functional group. Examples of suitable protecting groups for various functional groups are described in T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis. 2d. Ed.. John Wiley and Sons (1991) ; L. Fieser and M. Fieser, F eser and Fieser's Reagents for Organic Synthesis. John Wiley and Sons (1994) ; L. Paquette, ed. Encyclopedia of Reagents for Organic Synthesis. John Wiley and Sons (1995) .
The term "fused" whether preceded by the term "optionally" or not, refers to a structure wherein two distinct ring systems are joined together such that both rings share at least two common atoms. This can be envisioned as the replacement of a carbon-hydrogen or nitrogen-hydrogen bond on a ring atom with a carbon- carbon (from a second ring) or nitrogen-carbon (from a second ring) bond. For example, a cyclohexyl ring fused to a second cyclohexyl ring results in a decahydronaphthalene, a cyclohexyl ring fused to a piperidine ring results in a decahydroqumol e or decahydroisoqu olme, or a phenyl ring fused to a thiazole ring results in a oenzothiazole. The term "substituted", whether preceded by the term "optionally" or not, and substitutions contained in formulas of tnis invention, refer to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent. When more than one position in a given structure may be substituted with more than one substituent selected from a specified group, the substituents may be either the same or different at every position (for example,
2 2 the moiety -N(R ) (R )) . Typically, when a structure may be optionally substituted, 0-3 substitutions are preferred, and 0-1 substitutions is more preferred. Most preferred substituents are those which enhance protease inhibitory activity or intracellular antiviral activity in permissive mammalian cells or immortalized mammalian cell lines, or which enhance deliverability - 1 6 -
by enhancing solubility characteristics or enhancing pharmacokmetic or pharmacodynamic profiles as compared to the unsubstituted compound. Other more preferreα substituents include those used in the compounds shown Tables 1-5.
The term "pharmaceutically effective amount" refers to an amount effective in treating HIV infection in a patient either as monotherapy or n combination with other agents. The term "treating" as used herein refers to the alleviation of symptoms of a particu-ar disorder m a patient or the improvement of an ascertainable measurement associated with a particular disorder. Specifically, with respect to HIV, effective treatment using the compounds and compositions of tnis invention would result in an improvement in an HIV associated ascertainable measurement. The term "prophylactically effective amount" refers to an amount effective m preventing HIV infection in a patient. As used herein, the term "patient" refers to a mammal, including a human.
The term "pharmaceutically acceptable carrier or adjuvant" refers to a carrier or adjuvant that may be administered to a patient, together with a compounα of this invention, and which does not destroy the pharmacological activity thereof and is nontoxic when administered doses sufficient to deliver a therapeutic amount of the antiretroviral agent. As used herein, the compounds of this invention, including the compounds of formula I are defined to include pharmaceutically acceptable αerivatives or prodrugs thereof. A "pharmaceutically acceptable derivative or prodrug" means any pharmaceutically acceptable salt, ester, salt of an ester, or other derivative of a compound of this invention which, upon administration to a recipient, is capable of providing (directly or indirectly) a compound of this invention or an hibitorily active metabolite or residue thereof. Particularly favoreα derivatives and prodrugs are those that increase tne bioavailability of the compounds of this invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases. Examples of suitable acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycollic, lactic, salicylic, succmic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene- 2-sulfonιc and benzenesulfonic acids. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.
Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N- (C;L_4 alkyl) 4 salts.
The term "thiocaroamates" refers to compounds containing the functional group N-S02-0.
The compounds of this invention contain one or more asymmetric carbon atoms and thus occur as racemates and racemic mixtures, single enantiomers, diastereo eric mixtures and individual diastereomers. All such lsomeric forms of these compounds are expressly mcluded in the present invention. Each stereogenic carbon may be of the R or S configuration. Although the specific compounds exemplified in this application may be depicted in a particular stereochemical configuration, compounds having either the opposite stereochemistry at any given chiral center or mixtures thereof are also envisioned.
Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term "stable", as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a mammal or for use in affinity chromatography applications) . Typically, such compounds are stable at a temperature of 40°C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
The compounds of the present invention may be used in the form of salts derived from inorganic or organic acids. Included among such acid salts, for example, are the following: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camph- orsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, gluconepta- noate, glycerophosphate, hemisulfate, heptanoate, hexa¬ noate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, lactate, maleate, methanesulfonate, 2-naphthalenesulfonate, nicot ate, oxalate, pamoate, pectinate, persulfate, 3- phenylpropionate, picrate, pivalate, propionate, succmate, tartrate, thiocyanate, tosylate and undecanoate.
This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. The basic nitrogen can be quaternized with any agents known to those of ordinary skill in the art including, for example, lower alkyl halides, such as methyl, ethyl, propyl and butyl chloride, bromides and iodides; dialkyl sulfates including dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; and aralkyl halides including benzyl and phenethyl bromides. Water or oil-soluble or dispersible products may be obtained by such quaternization.
The compounds of this invention are those of formula I:
Figure imgf000021_0002
Figure imgf000021_0001
wherein each Z is
Figure imgf000022_0001
wherein any Z may be optionally fused with R ; each X and X' is independently selected from the group consisting of -C(O)-, -C(0)C(0)-, -S (0) - and -S(0)2; each Y and Y' is independently selected from the group consisting of -(C(R2)2)p-r -NR2-, - (C (R2) 2)p-M-, >C=C(R2)2, and -N(R2)-CH2-; each R is independently selected from the group consisting of hydrogen; R ; C]_-C6 alkyl; C2-C6 alkenyl; C2-C6 alkynyl; C3-C6 cycloalkyl optionally fused with
Figure imgf000022_0002
R ; C5-C6 cycloalkenyl optionally fused with R ; and where R 's are attached to adjacent atoms, the R 's together with their attached adjacent atoms form a carbocyclic or heterocyclic ring system which may be optionally fused with R ; where any member of R may be
2 optionally substituted by one or more R ;
2 each R is independently selected from hydrogen;
3 R ; Cλ-C6 alkyl; C2~C6 alkenyl; C2-C6 alkynyl; C3-C6 cycloalkyl optionally fused with R ; C5~C6 cycloalkenyl optionally fused with R ; and where two R 's are
2 attached to the same gemmal atom, the R 's together with their attached gemmal atom may form a spirocarbocyclic or spiroheterocyclic ring system;
2 where any member of R may be optionally substituted by one or more R,3" each R is independently selected from oxo, OR",
N(R9)2, N(R9)-X-R9, N(R9)-X-OR9, N(R9)-X-N(R9)2 , SR9, X-
R9, 0-X-N(R9)2, C(0)N(R9)2, halogen, N02, CN, COOR9 and
R6; each R is independently selected from from tne
9 9 9 9 6 group consisting of OR ; N(R )2; X-R ; C(0)N(R )2; R ; C^-Cg alkyl; C2-C4 alkenyl; C3-C6 cycloalkyl optionally fused with R ; C5~Cg cycloalkenyl optionally fused with R ; where any member of R may be optionally substituted by one or more groups independently selected from the group consisting of R 9 and R3; each R is independently selected from the group consisting of H, OH, 0 and R ; each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl may be optionally substituted with one or more groups selected from the group consisting of oxo, -OR 9, -R9, -N(R9) (R9),
-N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3;
7 each R is independently selected from the group consisting of hydrogen, OH and 0; p each R is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, and heterocyclyl;
9 each R is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heterocyclyl, aralkyl, carbocyclylalkyl and heterocyclylalkyl wherein any aryl, carbocyclyl or p heterocyclyl may be optionally fused with R and p wherein any member of R may be optionally substituted by one or more groups independently selected from the
8 8 P group consisting of -OR , -N(R )2, -CN, -N02, -X-R , -X-
N(R8)2 -C(0)OR8, -N(R8)-XNR8, and halogen; each Q is independently selected from CH and N; each M is independently selected from the group consisting of NH, -NR2-, -0-, -S-, -S(O)- and -S(0)2- each n is 1 or 2;
Figure imgf000024_0001
each p is independently 1 or 2; each q s independently 1, 2 or 3; and each G is independently selected from the group consisting of -NH-, -NR -, -0-, -S-, -S(O)-, S(0)2, -C(O)-, and -C(R2)2-.
Except where expressly noted to the contrary, the term "[variable] as defined for formula I" refers to the definitions shown directly above. In addition, where no reference is made to a particular definition for a given variable, the definition is to be taken as tnat defined for formula I shown directly above.
Preferred compounds of formula I are tnose wherein each Y and Y' is independently selected from the
2 2 2 group consisting of - (C (R ) 2) p-, -NR -, -(C(R )2)p-M-, and -N(R2)-CH2-; and
3 9 each R is independently selected from oxo, OR ,
N(R9)2, N(R9)-X-R9, N(R9)-X-OR9, SR9, X-R9, 0-X-N(R9)2,
C(0)N(R9)2, halogen, N02, CN, COOR9 and R6. Alternate preferred compounds of formula I are those having the structure of formula IA:
wherein
12 each R is independently selected from the group consisting of R ; Cι_-C6 alkyl optionally substituted with R ; C2-C6 alkenyl; C2-C6 alkynyl; C3-C6 cycloalκyI optionally fused with R ; C5-C6 cycloalkenyl optionally fused with R 6; where any memιbbe(r of R12 may be optionally ,2 substituted by one or more R
Preferred compounds of formula I are those wherein n is equal to 1; those having the structure of formula II:
Figure imgf000025_0001
and those having the structure of formula III
Figure imgf000025_0002
(III)
Also preferred are compounds accordmg to formula I wherein X is -C(O)- or -S(0)2 _ and
Y is -(C(R )2)p-M-; those wherein X is -C(0)- or -S(0)2- and Y is (-C(R2)2-)p; those wherein X is -C(0)-, -C(0)C(0)- or -S(0)2-; and
Y is -N(R2)- or -N(R2)-CH2-. An alternate object of this invention is a novel class of compounds represented by formula IV:
Figure imgf000026_0001
wherein:
X and X' are independently -C(O)- or -S(0)2-;
Y is -(C(R2)2)-M-, -(C(R2)2)p-r ~N(R2)- or -N(R2)-
CH2-; and each R 1, R2, R7, R4, p and M is independently as defined for formula I.
Another object of this invention is a novel class of compounds represented by formula V:
Figure imgf000026_0002
wherein:
X is -C(O)- or -S (0)2-;
Y is -(C(R2)2)-M-, -(C(R2)2)p-, -N(R2)- or -N(R2)-
CH2-;
R10 is 0 or H2; each R is independently H, OH or 0, wherein both
R ,11 are not simultaneously hydrogen; Z is a structure of formula VI:
Figure imgf000027_0001
wherein any structure of formula VI is optionally fused with an aryl, carbocyclic or heterocyclic ring and is optionally substituted with 1-3 substituents
2 independently selected from R (where in formula V, if
R is H2, a methylene s implied); and each R1, R2, R7, R4, R8, p, q, G, M, Q and X' is independently as defined for formula I.
Also preferred are those compounds having the structure of formula V, wherein
R10 and R11 are 0; compounds having the structure of formula V, wherein
R10 and R11 are 0; q is 1;
Figure imgf000027_0002
X' is -C(0)-; compounds having the structure of formula V, wherein
R10 and R11 are 0;
Figure imgf000027_0003
4
R is t-butylammo; compounds having the structure of formula V, wherein
Figure imgf000027_0004
X is -C(0)-; (R2)2)p-; and
Figure imgf000027_0005
compounds having the structure of formula V wherein X and X' is -C(O)-;
Figure imgf000028_0001
R is H2; and one R is H and one R is OH;
Also preferred are those compounds of formula V wherein
X and X' is -C(O)-;
Y is -(C(R2)2)-;
Figure imgf000028_0002
one R is H and one R is OH; and
2 R within the definition of Y is selected from
3 hydrogen, R or C^-C alkyl optionally substituted with R3; those compounds of formula V wherein
Figure imgf000028_0003
Y is -(C(R2)2)-; R7 is H; R10 is H2; one R is H and one R is OH; and
2 R within the definition of Y is selected from
9 hydrogen, -N(R )2 or heterocyclyl, which may be optionally benzofused, and wherein said heterocyclyl may be optionally substituted with one or more groups
9 9 selected from the group consisting of oxo, -OR , -R ,
-N(R9) (R9), -N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-
OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3; those compounds of formula V wherein X and X' is -C(O)-;
Figure imgf000028_0004
R10 is H2; one R is H and one R is OH; and
2 R within the definition of Y is selected from ;ne group consisting of:
Figure imgf000029_0001
Figure imgf000029_0002
Figure imgf000029_0003
Figure imgf000030_0005
Figure imgf000030_0001
Figure imgf000030_0002
Figure imgf000030_0003
HO
0- A
Figure imgf000030_0004
Figure imgf000031_0001
those compounds according to formula V wherein: X and X' is -C(O)-; Y is -(C(R2)2)-;
R is H;
Figure imgf000031_0002
one R is H and one R is OH; and
2 at least one R within the definition of Y is aryl optionally substituted with one or more groups selected
9 9 9 9 from the group consisting of oxo, -OR , -R , -N(R ) (R ),
9 9 9 9 9 9 9 -N(R ) -X-R , SR , -X-R , -0-X-N(R ) 2 > -R -OR , -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3; those compounds according to formula V wherein:
X and X' is -C(O) -;
Y is -(C(R2)2)-; R7 is H;
R10 is H2; one R is H and one R s OH; and
2 at least one R within the definition of Y is C1-C6
3 alkyl optionally substituted with R ; those compounds according to formula V wherein:
Figure imgf000031_0003
Y is (C(R'
Figure imgf000032_0001
R10 is H2;
Figure imgf000032_0002
2 at least one R within the definition of Y is C_-C_ alkyl optionally substituted with R ; and
3 at least one R within the definition of Y is pyridyl, triazolyl, oxazolyl, isoxazolyl, pyrimidyl, pyrazolyl, pyridazmyl, thiazolyl, imidazolyl, thienyl thiadiazolyl, oxadiazolyl, triazmyl or pyrazmyJ
3 wherein said R may be optionally substituted with 1-3 substituents selected from -OR , -R , -N(R ) (R ) ,
9 9 9 9 9 9 9
-N(R )-X-R , SR , -X-R , -0-X-N(R )2, -R -OR , -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3. those compounds accordmg to formula V wherein: X and X' is -C(O)-;
Figure imgf000032_0003
R10 is H2; one R 11 is H and one R11 is OH;
2 at least one R within the definition of Y is C^-Cg
3 aikyl optionally substituted with R ; and 3 R within the definition of Y is aryl optionally
9 substituted with 1-3 substituents selected from -OR ,
-R9, -N(R9) (R9), -N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, and
-CF3.
Also preferred are those compounds accordmg to any of the aforementioned preferred compounds of formula V wherein: R is benzyl; and Z is
Figure imgf000033_0001
H
those compounds according to any of the aforementioned preferred compounds of formula V wherein:
R is benzyl optionally substituted with 1-3 substituents selected from -OR9, -N(R9) (R9), SR9, -X-R9,
9 9 -R -OR , -CN, halogen, -N02, and -CF3; those compounds according to any of the aforementioned preferred compounds of formula V wherein:
R is benzyl optionally substituted with 1-3 substituents selected from -OR9, -N(R9) (R9), SR9, -X-R9,
9 9
-R -OR , -CN, halogen, -N02, and -CF3; and Z is
Figure imgf000033_0002
those compounds according to any of the aforementioned preferred compounds of formula V wherein R is benzyl optionally substituted with 1-3 substituents selected from the group consisting of OCH3, OH and NH2; those compounds according to any of the aforementioneα preferred compounds of formula V wherein R is benzyl optionally substituted with 1-3 substituents selected from the group consisting of 0CH3, OH and NH2 and wherein Z is
Figure imgf000034_0001
An alternate embodiment of this invention is compounds according to formula V, wherein:
Figure imgf000034_0002
each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected from the group consisting of oxo, -OR 9, -R9, -N(P9) (R9) ,
-N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, -CF3, -0- (CH2)q-R6,
-0-(CH2)q-0R , 2, 3-methylenedιoxy and 3,4- methylenedioxy; and each X, X', Y, Y', Z, R1, R2, R3, R4, R5, R7, R8, R9, Q,
M, n, r, p, q and G is independently as defined for formula I; and those compounds according to formula V, wherein: each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected
9 9 9 9 from the group consisting of oxo, -OR , -R , -N(R ) (R ),
9 9 9 9 9 9 9
-N(R )-X-R , SR , -X-R , -0-X-N(R )2, -R -OR , -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, -CF3, -0- (CH2) q-R6, g -0-(CH2)q-0R , 2, 3-methylenedιoxy and 3,4- methylenedioxy;
2
R within the definition of Y is selected from hydrogen, R or _-C^ alkyl optionally substituted with
3 R ; and each X, X', Y, Y\ Z, R1, R3, R4, R5, R?, R8, R9, Q, M, n, r, p, q and G is independently as defined for formula I . those compounds of formula V wherein
X and X' is -C(0)-;
Y is -N(R2)-;
Figure imgf000035_0001
R is h2; and one R is H and one R is OH; and those compounds of formula V wherein
X and X' is -C(O)-;
Figure imgf000035_0002
one R is H and one R11 is OH. Also preferred is the compound of formula I having the structure of formula IX:
Figure imgf000036_0001
(IX)
wherein
X is -C(O)- or -S(0)2-; and the compounds of formula IX wherein
X is -C(O)-;
Y is - (C(R2)2)-M-; and
7
R is H; and those compounds of formula IX wherein
Figure imgf000036_0002
Y is -N(R2)-; and
R is H; and those compounds of formula IX wherein X is -C(O)-; Y is -(C(R2)2A and R7 is H.
Also preferred are those compounds of formula I having the structure of formula XII:
Figure imgf000036_0003
(XII)
wherein
X and X' are independently -C(O)- or -S(0)2-; those compounds of formula I having the structure of formula XII, wherein
X and X' are independently -C(O)- or -S(0)2~; and 4 R is l-amino-2-hydroxyindanyl; and compounds of formula I having the structure of formula
4 XII, wherein R is 1 (S) -ammo-2 (R) -hydroxymdanyl .
Also preferred are the compounds according to formula I, having the structure of formula XIII:
Figure imgf000037_0001
wherein
X and X' are independently -C(0)- or -S(0)2-; compounds according formula I having the structure of formula XIII, wherein
X is -C(0)- or -S(0)2-;
X' is -C(0)-;
Y is -(C(R2)2)~ or -N(R2)-; and
Figure imgf000037_0002
compounds of formula I having the structure of formula XIII, wherein
X is -C(0)-;
Figure imgf000037_0003
Y is - (C(R2)2)-; and R7 is H; those compounds of formula XIII wherein X is -C(0)-; X' is -C(0)-;
Y is -(C(R2)2)-; R7 is H; and R within the definition of Y is selected from 3 hydrogen, R , or C]_-Cg alkyl optionally substituted with
R3; those compounds according to formula XIII wherein: X is -C(O)-;
X' is -C(0)-;
Y is -(C(R2)2)-;
R is H; and
2
R within the definition of Y is selected from
9 hydrogen, -N(R )2 or heterocyclyl, which may be optionally benzofused, and wherein said heterocyclyl may be optionally substituted with 1-3 groups selected
9 9 9 9 from the group consisting of oxo, -OR , -R , -N(R ) (R ) ,
9 9 9 9 9 9 9
-N(R )-X-R , SR , -X-R , -0-X-N(R )2, -R "OR , -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3; those compounds according to formula XIII wherein:
X is -C(O)-;
X' is -C(0)-;
Y is - (C(R2)2)-;
7 R is H; and
2 at least one R within the definition of Y is selected from the group consisting of:
Figure imgf000038_0001
3C ΎMe
Figure imgf000039_0001
Figure imgf000039_0002
Figure imgf000039_0003
Figure imgf000039_0007
Figure imgf000039_0004
Figure imgf000039_0005
V
Figure imgf000039_0006
H^ - 3
Figure imgf000040_0001
Figure imgf000040_0002
those compounds according to formula XIII wherein: X is -C(0)-;
Figure imgf000040_0003
Y is -(C(R2)2)-;
7
R is H; and at least one R within the definition of Y is aryl optionally substituted with one or more groups selectee 9 c o from the group consisting of oxo, -OR , -R , -N(R"1 (R"), -N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9~OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3; those compounds according to formula XIII wherein:
X is -C(O)-;
X' is -C(O)-;
Y is -(C(R2)2)-;
Figure imgf000041_0001
2 at least one R within the definition of Y is ^ - Q alkyl optionally substituted with R ; those compounds according to formula XIII wherein:
X is -C(O)-; X' is -C(O)-;
Y is -(C(R2)2)-;
Figure imgf000041_0002
at least one R within the definition of Y is pyπdyl, triazolyl, oxazolyl, isoxazolyl, pyrimidyl, pyrazolyl, pyridazmyl, thiazolyl, imidazolyl, thienyl thiadiazolyl, oxadiazolyl, triaz yl or pyraz y wherein said R may be optionally substituted with 1-3 substituents selected from -OR 9, -R9, -N(R9) (R9),
-N(R 9) -X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, or -CF3; those compounds according to formula XIII wherein:
X is -C(O)-;
X' is -C(O)-;
Y is -(C(R2)2)-; R7 is H; and R within the definition of Y is aryl optionally substituted with 1-3 substituents selected from -OR ,
-R9, -N(R9) (R9), -N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, or
-CF3; those compounds accordmg to any of the aforementioneα preferred compounds of formula XIII wherein: each R is benzyl; and
9 each R not withm the definition of Y is 2- hydroxymdanyl. those compounds according to any of the aforementioned preferred compounds of formula XIII wherein: each R is independently selected from benzyl optionally substituted with 1-3 substituents selected
9 9 9 9 9 9 9 from -OR , -N(R ) (R ), SR , -X-R , -R -OR , -CN, halogen, -N02, and -CF3; those compounds accordmg to any of the aforementioned preferred compounds of formula XIII wherein: each R is independently selected from benzyl optionally substituted with 1-3 substituents selected 9 9 9 9 9 9 from -OR , -N(R ) (R ), SR , -X-R , -R -OR , -CN, halogen, -N02, and -CF3; and
9 each R not within the definition of Y is 2- hydroxymdanyl; those compounds accordmg to any of the aforementioned preferred compounds wherein: each RI is independently selected from benzyl optionally substituted with 1-3 substituents selected from the group consisting of OCH3, OH and NH2; and those compounds accordmg to any of the aforementioned preferred compounds wherein: each RI is independently selected from benzyl optionally substituted with 1-3 substituents selected from the group consisting of 0CH3, OH and NH
9 eeaacchh RR nnoot withm the definition of Y is 2- hydroxymdanyl . Another embodiment is compounds according to formula XIII, wherein:
Figure imgf000043_0001
(XIII)
each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected from the group consisting of oxo, -OR 9, -R9, -N(R9) (R9),
-N(R 9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, -CF3, -0- (CH2)q-R°,
-0- (CH2)g-OR9 , 2, 3-methylenedιoxy and 3,4- methylenedioxy; and each X, X', Y, Y', Z, R1, R2, R3, R4, R5, R7, R8, R9, Q,
M, n, r, p, q and G is independently as defined for formula XIII .
Another embodiment is compounds accordmg to formula XIII, wherein:
Figure imgf000043_0002
(XIII)
wherein R withm the definition of Y is selected from hydrogen, R or Cχ-Z6 alkyl optionally substituted with R3; each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected
9 9 9 9 from the group consisting of oxo, -OR , -R , -N^R ) (R ),
9 9 9 9 9 9 9
-N(R )-X-R , SR , -X-R , -0-X-N(R )2, -R -OR , -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, -CF3, -O- (CH?) q-R6,
-0-(CH2)q-0R , 2, 3-methylenedιoxy and 3,4- methylenedioxy; and each X, X', Y, Y', Z, R1, R3, R4, R5, R7, R8, R9, Q, M, n, r, p, q and G is independently as defined for formula XIII.
Another embodiment is compounds of formula I having the structure of formula XIII, wherein
Figure imgf000044_0001
Y is -N(R2)-; and
Figure imgf000044_0002
compounds of formula I having the structure of formula XIII, wherein
Figure imgf000044_0003
Y is - (C (R2) 2)-; and
Figure imgf000044_0004
compounds of formula I having the structure of fcrmula
XIII, wherein
Figure imgf000044_0005
Y is -N(R2)-; and R7 is H.
In an alternate embodiment, preferred comDounds are those of formula V wherein
R10 is H2; and one R is H and one R is OH; and Z is selected from the group consisting of
and R is as defined in formula I; and those of formula V wherein Z is selected from the group consisting of
Figure imgf000045_0002
R is H2; and one R 11 is H and one R11 is OH.
Also preferred are those compounds of formula
V wherein X and X' is -C (0} - ;
Figure imgf000045_0003
one R 11 is H and one R11 is OH; and those compounds of formula V wherein
Figure imgf000045_0004
R is H ; and one R11 is H and one R11 is OH, and those compounds of formula V, wherein
X and X' is -C (0)-;
Figure imgf000046_0001
R7 is H;
R is H ; and one R is H and one R s OH, and the aforementioned compounds of formula V wherein Z is selected from the group consisting of:
Figure imgf000046_0002
2 and R is as defined in claim 1.
Also preferred are those compounds of formula V wherein X and X' is -C(O)-; Y is -(C(R2)2)-;
Figure imgf000046_0003
one R is H and one R is OH; and those compounds of formula V wherein X and X' is -C (0)-;
Y is -N(R2)-; R7 is H;
Figure imgf000046_0004
one R is H and one R is OH, and those compounds of formula V, wherein X and X' is -C(0) -;
Y is -(C(R2)2)-M-; M is 0; R7 is H;
R is H2; and one R 11 is H and one R11 is OH, and the aforementioned compounds of formula V wherein Z s selected from the group consisting of:
Figure imgf000047_0001
Also preferred are compounds of formula wherein:
Figure imgf000047_0002
4 Z is selected from the group consisting of -X'R ,
-NfRp-X'-R4, -NfR^-NfRp-X'-R4, and formula VI;
Figure imgf000047_0003
(VI) wherein any structure of formula VI is optionally fuseα with an aryl, carbocyclic or heterocyclic ring ana s optionally substituted with 1-3 members independently
2 selected from R ; and each X, X', Y, Y' R1, R2, R3, R4, R5, R6, R7, R8, R9, Q,
M, n, r, p, q and G is independently as defined for formula I.
Another embodiment of this invention relates to the process for preparing a compound of formula XIV:
Figure imgf000048_0001
XIV
wherein R and R are defined as in formula I, comprising the steps of:
(1) reacting a compound of formula XV:
Figure imgf000048_0002
wherein R is defined as in formula I, in an inert solvent, preferably an ethereal solvent such as diethyl ether or THF, with a base, preferably an alkali metal amide such as lithiumdiisopropylamide at a temperature between about -78 °C to about 25 °C;
(2) reacting the product of step (1) with an aldehyde R CHO followed by an optional treatment ' dehyrat g agent, preferably Martin's sulfurane dehydrating agent, wherein R is defined as m formula I to give a compound of formula XVI:
Figure imgf000049_0001
wherein R and R are defined as in formula I;
(3) reacting the product of step (2) m an inert solvent, preferably methanol, with hydrogen gas m the presence of an hydrogenation catalyst, preferably 10- palladium on carbon, followed by treatment with an anhydrous acid, preferably trifluoroacetic acid or 4N HCl in dioxane to give a product of formula XIV.
Another embodiment of this invention relates to the process for preparing a compound of formula XVII:
Figure imgf000049_0002
1 2 wnerein R and R are defined as in formula I, comprising the steps of:
(1) reacting a compound of formula XVIII: - 41
Figure imgf000050_0001
XVIII
1 2 wherein R and R are as defined in formula I, in an inert solvent, preferably DMF or THF, with a base preferably sodium hydride, then bromomethylacrylic acid at a temperature between about -78 °C to about 25 °C; (2) reacting the product of step (1) with ar. oxidizing agent, preferably ozone and if necessary a reductive work-up with a reducing agent such as dimethylsulfide;
(3) reacting the product of step (2) in an inert solvent, such as DMF, with thioproline t-butylamide and suitable amide-bond coupling reagents, preferably EDC,
HOBT and N-methylmorpholine, to give a product of formula XVII.
Another embodiment of this invention relates to the process for preparing a compound of formula XIX:
Figure imgf000050_0002
wherein R and r are defined as in formula I, comprising the steps of: 1) reacting a compound of formula XX
Figure imgf000051_0001
XX
wherein R ,1 i.s defined as in formula I and PG is a N- protecting group, such as those described in Greene and Wuts (infra) , preferably p-methoxybenzyl, an inert solvent, preferably THF, with a base, preferably lithiumdiisopropylamide at between about -78 °C to about 25 °C, then a bis-leaving group alkane of formula XXI:
Figure imgf000051_0002
wherein LG is selected from halo, preferably chloro or oαo, arylsulfonate esters, preferably tosyl, and alkylsulfonate esters, preferably mesyl, and r is defined as in formula I, to give a product of formula XXII:
Figure imgf000051_0003
wherein R and PG are defined as m formula XX and LG and r are defined as in formula XXI;
(2) reacting the product of step (1) in an inert solvent, preferably THF, with a base, preferably lithiumdiisopropylamide, at between about -78 °C to about 25 °C to give a product of formula XXIII:
Figure imgf000052_0001
wherein R is defined as in formula I and PG is a N- protecting group; (3) reacting the product of step (2) m an inert solvent with a reagent suitable for removal of the N- protecting group PG, such as those described in Greene and Wuts (inf,ra) . to give a compound of formula XIX.
In another embodiment, compounds of formula I with structures VII, VIII, IX, and X are preferred:
Figure imgf000052_0002
( VIII ) ( X )
where all definitions of variables for formula I apply.
2
Preferred R groups for formula I include:
C]_-Cg alkyl and alkenyl optionally substituted with R ;
2 where two R taken together form a spriocyclic ring and
C -Cg cycloalkyl or cycloalkenyl optionally fused with R6. Preferred compounds of this invention of formula I include the specific compounds contained i: Tables 1-5.
TABLE 1
Figure imgf000053_0001
Figure imgf000053_0002
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
n
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
- 6!
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
- 7!
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
I&BLE_2
Figure imgf000083_0001
Figure imgf000083_0002
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
15 -
Figure imgf000087_0001
16 -
Figure imgf000088_0001
57 -
Figure imgf000089_0001
18 -
Figure imgf000090_0001
19 -
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0002
TABLE 3
Figure imgf000100_0001
Figure imgf000100_0003
Figure imgf000101_0002
TAB E 4
Figure imgf000101_0001
Figure imgf000101_0003
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000104_0002
The preferred compounds of this invention are compound numbers (as in Tables 1-5) : 1, 2, 3, 4, 7, 8, 9, 13, 14, 16, 17, 18, 20, 23, 24, 25, 26, 32, 35, 38, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 62, 63, 72, 75, 76, 78, 80, 82, 83, 91, 92, 94, 95, 96, 101, 102, 109, 121, 122, 123, 124, 126, 127, 128, 129, 131, 132, 133, 134, 135, 137, 138, 140, 141, 145, 146, 147, 149, 150, 155, 156, 160, 161, 162, 164, 165, 170, 171, 175, 176, 177, 179, 180, 185, 186, 190, 191, 192, 194, 195, 200, 201, 208, 219, 220, 228 and 264. More preferred are compound numbers: 2, 7, 8, 9, 14, 18, 20, 25, 26, 32, 38, 45, 47, 48, 49, 50, 51, 53, 54, 62, 63, 72, 82, 83, 91, 92, 94, 95, 96, 123, 126, 140, 141, 219, 220, 228 and 264. Even more preferred are compound numbers: 7, 8, 9, 20, 45, 50, 51, 53, 54, 82, 83, 92, 94, 96, 219, 220, 228 and 264.
In an alternate embodiment, this invention also relates to novel methods for preparing compounds and intermediates of the following structures. One embodiment relates to a process
The compounds of this invention may be synthesized using conventional techniques. Advantageously, these compounds are conveniently synthesized from readily available starting materials. Although the syntheses of the compounds of this invention are known to those of skill in the art, the following general schemes are set forth to illustrate these methods. These schemes should not be viewed as limiting the scope of this invention m any way.
Using standard techniques, compounds of the present invention having the general formula I may be obtained as described in the following schemes:
SCHEME 1
Figure imgf000106_0002
raductiv* amination
Figure imgf000106_0001
Figure imgf000106_0003
El EV
1 ddeapprorottact Mg / MeOH
2 base/ heat
R1 — OH
1 O-activation
H -^ " 2 nucleophile
R NH ς»
O H O o
Ell EVII EVI
Figure imgf000106_0004
E3
SCHEME 1 (confd)
Figure imgf000107_0001
1. athyldiβthyl 1. MβONHMe / EDCI phσsphoranyl
2. reduce mβthanasulfonate
3. TMSCHjMgBr n-BuLi
4. BF3 atharate
2. reduction
5. AcSH / hv
6. Cl2 / AcOH / HCl
P - ^^s-OMe ^^"*e-CI o- -o o'- "O
Ellla Elllb
1. daprotact
2. cyclize
Figure imgf000107_0002
EIV
1. protect
2. base. "E"
3. daprotact
R'
:- -NH
" o'-s-'o
E2
SCHEME 2
Figure imgf000108_0001
EVIII
Figure imgf000108_0002
7 ΓΛ
2 /- R N. .NH
R N NH 2N^, R1
Y o"-s-"o
O
E5 o
E4
E6
SCHEME 3
Figure imgf000109_0001
Route A Route B
EIX
Figure imgf000109_0002
E7
1. daprotact
2. base / heat
.OTBDMS
*W H
EX
1. TBDMSCl
2. deprotect _.
Route C 3. base. ^O e
R A2
%O>H
SCHEME IV
Figure imgf000110_0001
* E8
(Z - E1 - E7)
R"'
Figure imgf000110_0002
EXIa EXIb
Figure imgf000110_0003
1 allyl bromide
2 epoxidee
Figure imgf000111_0004
Figure imgf000111_0001
SCHEME 6
R ' 1 O - protect 2 i... b caasee /' 'E c" 3 deprotect
Figure imgf000111_0002
Figure imgf000111_0003
Methods for producing the compounds of this invention are well known in the art of organic synthesis. Several intermediates are commercially available, e.g. from Aldrich Chemical Company, Inc., Milwaukee, Wl. The synthesis of heterocycles E1-E6 (Schemes 1 and 2) begins with any protected amino aldehyde, the preparations for which are well known in the art from suitably protected amino acids, esters or alcohols. In the case of the this intermediate, transient protection of the amino group may be accomplished by means known in the art (see, e.g. T.W, Greene and P.G.M. Wuts "Protective Groups in Organic Synthesis", Second Edition, pp. 309-405 ©1991 John
Wiley and Sons, Inc. New York, NY and E. Gross and J. Memhofer "The Peptides, Vol. 3: Protection of Functional Groups m Peptide Synthesis" pp. 3-88; ©1981 Academic Press, Inc. New York, NY) . Carbamates such as Boc, Fmoc, Alloc and Cbz are particularly convenient protecting groups, the introduction and removal of are described in the above references.
The synthesis of El is illustrated m Scheme 1. The protected ammo aldehyde is treated with an alpha substituted or alpha, alpha disubstituted amino ester under typical reductive amination conditions well known m the art, such as sodium cyanoborohydride in a solvent mixture of DMF/Acetic acid. The resulting compound El is then deprotected and free based with either a tertiary amme base or potasiu carbonate in methanol to effect cyclization to form EII The resulting secondary amme may the be protected with groups (detailed in the references above) such as benzyl or t-butyloxycarbonyl (Boc) utilizing conditions well known in the art to form analogs of El. Preparation of E2 is achieved by reaction of a starting aldehyde with ethyl diethylphosphoranylmethanesulfonate and subsequent reduction of the double bond (see: Gennari et al., Angew. Chem. Int. Ed. Engl., 33, pp. 2067-69 (1994)) to yield compound Ellla. Cyclization may then be achieved by deesterification and activation of the sulfonate moiety as described in Gennari, followed by deprotection of the nitrogen protection group to yield the cyclized product EIV. Alternatively, an ammo acid may be converted to compound Elllb using standard synthetic methods illustrated in Scheme 1. Compound Elllb can be cyclized to afford compound EIV. Compound EIV may then be N-protected, for example, the presence of Boc anhydride and DMAP (see: Flynn et al . , - I l l -
orς cn π 46, pp. 2424~26 (1983)), and treated with a non-nucleophilic base such as LDA or hexamethyldisilazane to generate the anion at the center alpha to the S02 moiety. This anion may then be quenched with a variety of electrophiles and subsequently deprotected to form the desired analogs of E2. Alternatively, this anion may be quenched with an aldehyde to form (after subsequent dehydration, i.e., an aldol-type condensation) an exo-methylene compound which may then be reduced {i.e., hydrogenation) to form tne desired analogs of E2. Analogously, preparation of E3 results from a Wittig reaction using methyl (triphenylposphoranylidene) acetate followed by simultaneous reduction of the double bond and cyclization using magnesium metal methanol (Wei et al., Tetrahedron Lett., 34(28), pp. 4439-42 (1993)) . A similar N-protection, deprotonation, quench and N- deprotection scheme, or condensation-reduction scheme, as described in the preparation of E2, results m desired analogs of E3. Alternatively, E3 may be prepared from commercially available EVI. The hydroxyl group may be activated using commonly available reagents such as methanesulfonyl chloride or para- toluenesulfonyl chloride m the presence of a tertiary amine base. The addition of a nucleophile to displace the mesylate or tosylate yields EVII (Ackermann et al., Helv. Chim. Acta, 73, pp. 122-32 (1990)) which may be treated as described above to obtain E3.
Methods for the preparation of compounds E4- E6 are also well known in the art and stem from readily available protected ammo aldehydes. Treatment of these aldehydes with a variety of amines under reductive amination conditions well known in the art, such as sodium cyanoborohydride using DMF/Acetic acid as a solvent mixture, followed by deprotection of the primary amme yields diamine EVIII. Intramolecular cyclization with a variety of activated carbonyl, dicarbonyl or sulfuryl equivalents m the presence of a tertiary amme base yields compounds E4-E6. Examples of activating reagents include but are not limited to carbonyldiimidazole, phosgene, sulf ryldichloride, sulfuryldiimidazole, sulfonyl dumide, and oxalyl chloride. Methods leading to the production of analogs of compound E7 are also known m the art (McManus et al., J. Med. Chem., 8, pp. 766-76 (1965)) . Scheme 3 exemplifies several potential routes to the synthesis of compound E7. Any protected ammo alcohol may be deprotonated to form the alkoxide which may be reacted with a substituted alpha bromo ester to form ether EIX
(route A) . Alternatively (route B) , EIX may be formed from activation of a protected ammo alcohol with, for example, methanesulfonyl chloride or para- toluenesulfonyl chloride the presence on a tertiary amme base and subsequent addition of a nucleophile such as an alkoxide from an alpha hydroxy acid to displace mesylate or tosylate to yield EIX. Compound EIX can then be deprotected, free based with a tertiary amme base or potassium carbonate in methanol, and heated to effect cyclization to form E7. Alternatively (route C) , E7 may be prepared from a protected ammo alcohol by protection of the hydroxyl group with, for example, t-butyldimethyl silyl chloride/imidazole to afford the silyl ether. Subsequent nitrogen αeprotection and acylation with a alpha bromo acid in the presence of any number of available coupling agents (for example dicylcohexylcarbodiimide, other related carbodiimide reagents or isobutyl chloroformate) or acylation with an alpha bromo acid chloride provides compound EX. Desilylation using, for example, tetrabutylammonium formate THF followed by formation of the alkoxide with base affords cyclization to E7. Alternatively, E7 may be prepared from the correspondmg a-methylene compound (i.e., both PJ are H in E7, the nitrogen may be protected if necessary) oy a multiple deprotonation-alkylation sequence to give an
2 E7 wherein each R is inserted in an independent
2 alkylation step and each R may be attached to form a spirocyclic product (i.e., alkylation with a dihaloalkane) .
Schemes 4-6 describe methods for converting the cyclic compounds E1-E7 into compounds of this invention. For example, compounds of the type Z, exemplified by compounds E1-E7, may be deprotonated and reacted with a functionalized epoxide to generate the desired compounds as described m Scheme . Several of the described epoxides are readily synthesized via methods well known in the art (Maligres et al,, Tetrahedron Lett., 36, pp. 2195-98 (1995)) . Optionally, further modification of the compounds may be performed subsequent to epoxide opening using reactions and materials well known in the art. For example, subsequent to epoxide opening utilizing example EXIb deprotection of the carbamate allows further modification of the unmasked amme.
Alternatively, as shown in Scheme 5, compounds EZ may be converted to the desired products m a more stepwise fashion. Compounds EZ may be αeprotonated using, for example, sodium hydride in DMF and treated with a three carbon based epoxide to generate epoxide EXII. Examples of such reagents include, but are not limited to, eDibro ohyαrm, epichlorohydrin and glycidyl tosylate. Several other potential methods for preparing compounds of the type EXII are well known in the art, for example, the anion of Z may be reacted with allyl bromide or allyl iodide to form an allyl intermediate, which may subsequently be oxidized to form the desired epoxide. Several epoxidation conditions for the generation of either racemic or chiral epoxides are well known in the art. Epoxide EXII may then treated with an amme and susequently carbonylated or sulfonated using activated species well known the art to generate final compounds of the type E9. Alternatively EXII may be reacted with a functionalized secondary amme followed
2 by optional manipulation of R to produce compounds of the type E10. One example of such manipulation is reaction of EXII with the known Boc piperazine EXIII (Dorsey et al . , J. Med. Chem. ,37, pp. 3443-51 (1994) ) . Subsequent to epoxide opening, the Boc group may be removed and the unmasked secondary amme may be further manipulated by reaction with various electrophiles to form the desired product.
Scheme 6 describes a method for introduction of electrophiles into comounds of the type EXIV. Said compounds may be protected with a variety of protecting groups, for example t-butyldimethylsilyl triflate, to mask the secondary hydroxyl group followed by treatment with a non-nucleophilic base such as lithium dusopropylamide or hexamethyldisilyzane to generate the anion alpha to the carbonyl. Various electrophiles may then be added to substitute the position alpha to the carbonyl, or alternatively an aldol-type condensation-reduction scheme may be employed. Deprotection of the secondary hydroxyl then yields the desired υroduct. As can be appreciated by the skilled artisan, the above synthetic schemes are not intended to comprise a comprehensive list of all means by which the compounds described and claimed in this application may be synthesized. Further methods will be evident to those of ordinary skill in the art.
Moreover, the determination of the optimum overall scheme, as well as the choice of reagents and reactions used to carry out the various steps in a given scheme will be based upon factors that are readily apparent to those of skill the art. These factors include the identity of the compound to be produced, the efficiency of the individual steps and schemes in producing that compound terms of overall yield, time, and cost and availability of reagents. It will therefore be apparent that some routine experimentation may be required in determining the optimum scheme to produce certain compounds of this invention. It should be understood that the compounds of this invention may be modified by appending appropriate functionalities to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system) , increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion. The compounds of this invention are characterized by a superior ability to inhibit protease activity and viral replication, particularly aspartyl protease activity. These compounds are especially well suited for inhibiting HIV aspartyl protease. We believe that this activity is due to specific steric and electronic interactions between the protease and compounds of this invention. This belief stems from our analysis of the structural basis for the activity of compounds of this invention, in view of the known crystal structures of HIV protease and bound inhibitors, such as the structure reported in Miller et al. "Structure of Complex of Synthetic HIV-1 Protease with a Substrate-Based Inhibitor at 2.3 A Resolution", Science, vol. 246, pp. 1149-1152 (1989), which is incorporated herein by reference, as well as structures determined m our laboratories.
The novel compounds of the present invention are excellent ligands for aspartyl proteases, particularly HIV-1 and HIV-2 proteases. Accordingly, these compounds are capable of targeting and inhibiting late stage events HIV replication, i.e., the processing of the viral polyprotems by HIV encoded proteases. Such compounds inhibit the proteolytic processing of viral polyprotem precursors by inhibiting aspartyl protease. Because aspartyl protease is essential for the production of mature virions, inhibition of that processing effectively blocks the spread of virus by inhibiting the production of infectious virions, particularly from chronically infected cells. Compounds accordmg to this invention advantageously inhibit the ability of the HIV-1 virus to infect immortalized human T cells over a period of days, as determined by an assay of extracellular p24 antigen — a specific marker of viral replication.
Other anti-viral assays have confirmed the potency of these compounds.
The compounds of this invention may be employed in a conventional manner for the treatment of viruses, such as HIV and HTLV, which depend on aspartyl proteases for obligatory events in their life cycle. Such methods of treatment, their dosage levels and requirements may be selected by those of ordinary skill in the art from available methods and techniques. For example, a compound of this invention may be combined with a pharmaceutically acceptable ad uvant for administration to a virally-mfected patient in a pharmaceutically acceptable manner and m an amount effective to lessen the severity of the viral infection or to alleviate pathological effects associated with HIV infection or immunosuppression such as opportunistic infections or various cancers, tumors, CMV retmitis, Candida infections, maternal fetal transmission, and AIDS related dementia, .
Alternatively, the compounds of this invention may be used in prophylactics and methods for protecting individuals against viral infection durmg a specific event, such as childbirth, or over an extended period of time. The compounds may be employed in such prophylactics either alone or together with other antiretroviral agents to enhance the efficacy of each agent. As such, the novel protease inhibitors of this invention can be administered as agents for treating or preventing HIV infection in a mammal.
The compounds of formula I, especially those having a molecular weight of less than about 700 g/mole, may be readily absorbed into the bloodstream of mammals upon oral administration. Compounds of formula I naving a molecular weight of less than about 600 g/mole and aqueous solubility of greater than or equal to 0.1 mg/mL are most likely to demonstrate high and consistent oral availability. This surprisingly impressive oral availability makes such compounds excellent agents for orally-administered treatment and prevention regimens against HIV infection.
The compounds of this invention may be administered to a healthy or HIV-infected patient either as a smgle agent or in combination with other anti-viral agents which interfere with the replication cycle of HIV. By administering the compounds of this invention with other anti-viral agents which target different events in the viral life cycle and which target different viral substrams with varying susceptability to specific agents, the therapeutic effect of these compounds is potentiated. For instance, the co-administered anti-viral agent can be one which targets early events in the life cycle of the virus, such as cell entry, reverse transcription and viral DNA integration into cellular DNA. Anti-FIV agents targeting such early life cycle events lrclude, didanosme (ddl), dideoxycytidine (ddC) , d4T, zidovudme (AZT) , 3TC, 935U83, 1592U89, 524W91, polysulfated polysaccharides, sT4 (soluble CD4) , ganiclovir, trisodium phosphonoformate, eflornith e, ribavirm, acyclovir, alpha interferon and tri- methotrexate. Additionally, non-nucleoside inhibitors of reverse transcriptase, delavirdine (U90) or nevirapme, may be used to potentiate the effect of the compounds of this invention, as may viral uncoatmg inhibitors, inhibitors of trans-activatmg proteins such as tat or rev, or inhibitors of the viral mtegrase. Combination therapies accordmg to th s invention exert an additive or synergistic effect inhibiting HIV replication because each component agent of the combination acts on a different site of HIV replication or on different strains of virus present in an infectious population. The use of such combination therapies may also advantageously reduce the dosage of a given conventional anti-retroviral agent which would be required for a desired therapeutic or prophylactic effect, as compared to when that agent is administered as a monotherapy. Such combinations may reduce or eliminate the side effects of conventional smgle anti- retroviral agent therapies, while not interfering with the anti-retroviral activity of those agents. These combinations reduce potential of resistance to smgle agent therapies, while minimizing any associated toxicity.
Advantages of combining HIV protease inhibitors may include viral population effects, whereby certain members of a virus population which show reduced sensitivity to one protease inhibitor may be fully sensitive to another inhibitor or may in fact have enhanced sensitivity to the second inhibitor. Alternatively or in addition, administration of two or more different inhibitors may be used to reduce specific toxicities associated with a smgle agent. This advantage of combination therapy also applies to co-admmistration of the protease inhibitor of this invention with other antiviral agents. Alternatively or in addition, co-administration of more than one protease inhibitor may lower the rate of metabolic activation of the compounds of this invention, for instance, by inhibiting enzymatic systems such as cytochrome P450/ or esterases or the like. In particular, co-administration of compounds of this invention with protease inhibitors such as ritonavir or other agents such as ketoconazole, grapefruit juice and antiulcer medications such as H2-blockers, which inhibits cytochrome P450 3A4, may advantageously enhance their biological half-life.
These combinations may also increase the efficacy of the conventional agent without increasing the associated toxicity. Compounds of this invention in combination with other anti-HIV agents may act in an additive or synergistical manner m preventing the replication of HIV in human T cells. Preferred combination therapies include the administration of a compound of this invention with AZT, ddl, ddC, d4T,
3TC, 935U83, 1592U89, 524W91 or a combination thereof.
Alternatively, the compounds of this invention may also be co-administered with other HIV protease inhibitors such as VX-478 (Vertex, also known as 141W94 (Glaxo-Wellco e) and KVX-478 (Kissei) ) , saqumavir (Ro 31-8959, Roche), indinavir (L-735,524, Merck) ), ritonavir {ABT 538, Abbott), nelfmavir (AG 1343, Agouron), palinavir (Bila 2011 BS) , U-103017 (Upjohn), XM 412 (DuPont Merck), XM 450 (DuPont Merck), BMS 186318 (Bristol-Meyers Squibb), CPG 53,437 (Ciba Geigy), CPG 61,755 (Ciba Geigy), CPG 70,726 (C oa Geigy), ABT 378 (Abbott), GS 3333 (Gilead Sciences), GS 3403 (Gilead Sciences) , GS 4023 (Gilead Sciences), GS 4035 (Gilead Sciences), GS 4145 (Gilead Sciences), GS 4234 (Gilead Sciences), and GS 4263 (Gilead Sciences) or prodrugs of these or related compounds to increase the effect of therapy or prophylaxis against various viral mutants or members of HIV quasi species.
We prefer administering the compounds of this invention as single agents or in combination with retroviral reverse transcriptase inhibitors, such as nucleoside derivatives, or other HIV aspartyl protease inhibitors, including multiple combinations comprising from 3-5 agents. We believe that the co-admmi tration of the compounds of this invention with retroviral reverse transcriptase inhibitors or HIV aspartyl protease inhibitors may exert a substantial additive or synergistic effect, thereby preventing, substantially reducing, or completely eliminating viral replication or infection or both, and symptoms associated tnerewith. Particularly preferred is administration of a combination of a compound of formula I, 3TC and zidovud e (AZT) . Also preferred are administrations of combinations of a compound of formula I and 1592U89, or of compounds of formula I with VX-478, optionally with one or more reverse transcriptase inhibitors, paarticularly, AZT, 3TC and 1592U89.
The compounds of this invention can also be administered in combination with immunomodulators and immunostimulators (e.g., bropirimme, anti-human alpha interferon antibody, IL-2, GM-CSF, interferon alpha, diethyldithiocarbamate, tumor necrosis factor, naltrexone, tuscarasol, and rEPO) ; and antibiotics (e.g., pentamid e lsethiorate) to prevent or combat infection and disease associated with HIV infections, such as AIDS, ARC and HIV-associated cancers.
When the compounds of this invention are administered in combination therapies with other agents, they may be administered sequentially or concurrently to the patient. The additional agents may be administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those agents may be part of a smgle dosage form, mixed together with the compounds of this invention in a single composition. The pharmaceutical compositions accordmg to this invention may comprise a combination of an aspartyl protease inhibitor of this invention and one or more therapeutic or prophylactic agents.
Although this invention focuses on the use of the compounds disclosed herein for preventing and treating HIV infection, the compounds of this invention can also be used as inhibitory agents for other viruses which depend on similar aspartyl proteases for obligatory events in their life cycle. These viruses include other AIDS-like diseases caused by retroviruses, such as simian immunodeficiency viruses, HTLV-I and HTLV-II. In addition, the compounds of this invention may also be used to inhibit other aspartyl proteases, such as renin, pepsin, cymosin, RSV protease, AMV protease, SIV protease and FIV protease, and in particular, other human aspartyl proteases, including renin, and aspartyl proteases that process endothelin precursors.
Pharmaceutical compositions of this invention comprise any of the compounds of the present invention, and pharmaceutically acceptable salts thereof, with any pharmaceutically acceptable carrier, adjuvant or vehicle. Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-α-tocopherol polyethyleneglycol 1000 succ ate, surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, polyethyleneglycol polymers such as PEG-400, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamme sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. Cyclodextrins sucn as α-, β-, and γ-cyclodextnn, or chemically modified derivatives such as hydroxyalkylcyclodextrms, including 2- and 3-hydroxypropyl-β-cyclodextrms, or other solublized derivatives may also be advantageously used to enhance delivery of compounds of formula I. The pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vagmally or via an implanted reservoir. We prefer oral administration or administration by injection. The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, mtracutaneous, intravenous, intramuscular, mtra- articular, intrasynovial, mtrasternal, mtrathecal, mtralesional, and mtracranial injection or infusion techniques.
The pharmaceutical compositions may be m the form of a sterile mjectable preparation, for example, as a sterile mjectable aqueous or oleaginous suspension. This suspension may be formulated accordmg to techniques known m the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile mjectable preparation may also be a sterile mjectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example, as a solution in 1, 3-butanedιol. Among the acceptable vehicles and solvents that may be employed are mannitol, water. Ringer's solution and lsotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful m the preparation of mjectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as carboxymethyl cellulose or similar dispersing agents which are commonly used m the formulation of pharmaceutically acceptable dosage forms such as emulsions and or suspensions. Other commonly used surfactants such as Tweens and Spans and/or other similar emulsifying agents or bioavailability enhancers which are commonly used m the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation. The pharmaceutical compositions of this invention may be orally administered in any ora] ly acceptable dosage form including, but not limited to, hard or soft gelatin capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration a capsule form, useful diluents include lactose and αried corn starch. When aqueous suspensions and/or emulsions are administered orally, the active ingredient may be suspended or dissolved in an oily phase combined with emulsifying and/or suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
The pharmaceutical compositions of this invention may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore w ll melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application. For application topically to the skin, the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolveα in a carrier with suitable emulsifying agents. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved m a carrier. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. The pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or m a suitable enema formulation. Topically-transdermal patches are also mcluded m this invention.
The pharmaceutical compositions of thi s invention may be administered by nasal aerosol or inhalation. Such compositions are prepared accordmg to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizmg or dispersing agents known in the art.
Dosage levels of between about 0.01 ana about 100 mg/kg body weight per day, preferably between about 0.5 and about 75 mg/kg body weight per day of the active ingredient compound are useful in the prevention and treatment of viral infection, including HIV infection. Typically, the pharmaceutical compositions of this invention will be administered from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. A typical preparation will contain from about 5* to about 95% active compound (w/w) . Preferably, such preparations contain from about 20% to about 80q active compound. Upon improvement of a patient's condition, a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
As the skilled artisan will appreciate, lower or higher doses than those recited above may be required. Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of tne specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the infection, the patient's disposition to the infection and the judgment of the treating physician.
The compounds of this invention are also useful as commercial reagents which effectively bind to aspartyl proteases, particularly HIV aspartyl protease. As commercial reagents, the compounds of this invention, and their derivatives, may be used to block proteolysis of a target peptide or may be derivatized to bind to a stable resin as a tethered substrate for affinity chromatography applications. For example, a compound of formula I may be tethered to an affinity column to purify recombinantly produced HIV protease. Derivatization of the compounds of this invention to produce affinity chromatography res s and the methods used to purify proteases using such resms are well known and with the skill of the art. These and other uses which characterize commercial aspartyl protease inhibitors will be evident to those of ordinary skill in the art. (See: Rittenhouse, J. et al. Biochem.
Biophvs. Res. Commun. 171, p. 60 (1990) and Heimbach, J.C. et al. Ib d 164, p. 955 (1989)) .
In order that this invention be more fully understood, the following examples are set forth. These examples are for the purpose of illustration only and are not to be construed as limiting the scope of the invention any way.
General Materials and Methods
All temperatures are recorded in degrees Celsius. Thin layer chromatography (TLC) was carried out using 0.25 mm thick E. Merck silica gel 60 F25 plates and elution with the indicated solvent system. Detection of the compounds was carried out by treating the plate with an appropriate visualizing agent, such as 10% solution of phosphomolybdic acid in ethanol or a 0.1% solution of nmhydrin in ethanol, followed by heating, and/or by exposure to UV light or iodine vapors when appropriate. Thick layer silica gel chromatography was also carried out using E. Merck 60 F25 plates ("prep plates") of 0.5, 1.0, or 2.0 mm thickness. Following development of the plate, the band of silica containing the desired compound was isolated and eluted with an appropriate solvent. Analytical HPLC was carried out using a Water's Delta Pak, 5 μM silica, C18 reversed-phase column, 3.9 mm ID x 15 cm L with a flow rate of 1.5 mL/mm using the following table:
Mobile phase: A = 0.1% CF3C02H m H20
B = 0.1% CF3C02H in CH3CN Gradient: T = 0 min., A (95%), B (5%) T = 20 min., A (0%), B (100%)
T = 22.5 mm., A (0%), B (100Ϊ)
Preparative HPLC was also carried out using C18 reversed-phase media. HPLC retention times were recorded in minutes. NMR spectral data was recorded using a Bruker AMX500, equipped with either a reverse or QNP probe, at 500 MHz, and was taken in the indicated solvent.
We have measured the inhibition constants of each compound against HIV-1 protease using the method described essentially by M.W. Pennmgton et al.,
Peptides 1990, Giralt, E. and D. Andreu, Eds., Escom, Leiden, Netherlands (1991); and the method described essentially by Partaledis et al., J. Virol .. 69, pp. 5228-35 (1995) . Compounds of invention were tested for their antiviral potency in several virological assays. In the first assay, the compounds were added as a solution in di ethylsulfoxide (DMSO) to a test cell culture of CCRM-CEM cells, a stram of CD4+ human T-cell lymphoma cells, previously acutely infected with HIVIIIb using standard protocols (see Meek, T. D. et al., "Inhibition of HIV-1 protease in infected T-lymphocytes by synthetic peptide analogues". Nature, 343, p. 90 (1990) . The effect of the compounds on inhibiting the replication of the virus was measured by determining the HIV extracellular p24 antigen concentration using a commercial enzyme immunoassay (obtained from Coulter Corporation, Hialeah, FL) . Antiviral activity may also be measured in a separate assay in MT4 cells. Antiviral HIV activity and compound-induced cytotoxicity were measured m parallel by means of a propidium iodide based procedure in the human T-cell lymphotropicvirus transformed cell line MT4. Aliquots of the test compounds were serially diluted in medium (RPMI 1640, 10% fetal calf serum (FCS) , and gentamycin) in 96-well plates (Costar 3598) using a Cetus Pro/Pette. Exponentially growing MT4 cells were harvested and centrifuged at 1000 rpm for 10 minutes in a Jouan centrifuge (model CR 4 12) . Cell pellets were resuspended in fresh medium (RPMI 1640, 20% FCS, 20% IL-2, and gentamycin) to a density of 5 x 105 cells/ml. Cell aliquots were infected by the addition of HIV-1 (strain IIIB) diluted to give a viral multiplicity of infection of 100 x TCID50. A similar cell aliquot was diluted with medium to provide a mockmfected control. Cell infection was allowed to proceed for 1 hour at 37 °C in a tissue culture incubator with humidified 5% C02 atmosphere. After the 1 hour incubation the virus/cell suspensions were diluted 6-fold with fresh medium, and 125 μl of the cell suspension was added to each well of the plate containing prediluted compound. Plates were then placed m a tissue culture incubator with humidified 5% C02 for 5 days. At the end of the incubation period, 27 ul of 5% Nonιdet-40 was added to each well of the incubation plate. After thorough mixing with a Costar multitip pipetter, 60 ul of the mixture was transferred to filter-bottomed 96-wellplates. The plates were analyzed in an automated assay instrument (Pandex Screen Machine, Baxter Biotechnology Systems) . The assay makes use of a propidium iodide dye to estimate the DNA content of each well. The antiviral effect of a test compound is reported -as an IC5o, i.e. the inhibitory concentration that would produce a 501 decrease in the HIV induced cytopathic effect. This effect is measured by the amount of test compound required to restore 50% of the cell growth of HIV- mfected MT-4 cells compared to unmfected MT-4 cell controls.
References:
1. Averett, D.R. 1989. Anti-HIV compound assessment by two novel high capacity assays. J. Virol. Methods 23: 263-276.
2. Schwartz, 0., et al . 1988. A rapid and simple colorimetric test for the study of anti-HIV agents. AIDS Res, and Human Retrovi uses. 4 (6) : 441-447.
3. Daluge, S.M., et al . 1994. 5-chloro-2 ' , 3 ' - dedeoxy-3 ' fluorouridme (935U83), a selective anti- human immunodeficiency virus agent with an improved metabolic and toxicological profile. Aαen s and Chemother.. 38 ( ): 1590-1603.
4. Dornsife, R.E., et al. 1991. Anti-human immunodeficiency virus synergism by zidovudme (3'- azidothymidine) and didanosine (dideoxymosme) contrasts with their additive inhibition of normal human marrow progenitor cells. Antim cro. Agents and Chemother.. 35(2): 322-328.
Depending on the cell type and the desired readout, syncytia formation, reverse-transcriptase (RT) activity, or cytopathic effect as assayed by a dye uptake method may also be used as readouts of antiviral activity. See H. Mitsuya and S. Broder, "Inhibition of the in vitro mfectivity and cytopathic effect of human T-lymphotropic virus type III/lymphoadenopathy- associated virus (HTLV-III/LAV) by 2', 3'- dideoxynucleosides", Proc. Natl. Acad. Sc . USA, vol. 83, pp. 1911-1915 (1986).
Insofar as the compounds of this invention are able to inhibit the replication of the HIV virus m CD4 cells of human lineage, they are of evident clinical utility for the treatment of HIV infection. These tests are predictive of the compounds ability to inhibit HIV protease __u. vivo.
Synthetic Examples
Example 1
Figure imgf000135_0001
N- (t-butoxycarbonyl) -L-phenylalaninol;
251.3 g/Mol 10. Og 39.8 mmol DMSO 78 g/Mol 3.80mL 49.0 mmol oxalyl chloride 126.9 g/Mol 3.82mL 43.8mmol triethylamine 101 g/Mol 23.0mL 160mmol methylene chloride 200 mL
The oxalyl chloride was added dropwise to a solution of DMSO in methylene chloride at -78 °C. After stirring for 10 minutes, the alcohol was added as a solution in methylene chloride. The reaction was then stirred at -78 °C for 45 minutes. At this time the triethylamine was added and a white precipitate formed. The reaction was then stirred 45 minutes at -78 °C and 45 minutes at 0 °C. The reaction was then quenched by the addition of a solution of 90g of citric acid in 300 mL of water. The organic portion of the reaction was then washed by (2 x 80 mL) of both saturated sodium bicarbonate and brine. The combined organic layers were then dried over sodium sulfate, filtered and concentrated in vacuo to leave a white solid. The aldehyde was then used without further purification the reductive amination.
B ,
Figure imgf000136_0001
1
allyl amine 57 g/Mol 6.0 L 160 mmol aldehyde est. 39.8 mmol sodium cyanoborohydride 62.8g/Mol 4.Og 6.4 mmol
D F 180 mL acetic acid (glacial) 1.8 mL
The aldehyde of Example 1A was dissolved in 180 mL of DMF at 25 °C. This was followed by addition of the aldehyde and 1.8 mL of acetic acid respectively. After 2 hours sodium cyanoborohydride was added, as a solid. The reaction was then stirred at 25 °C for 12 hours.
The reaction was then quenched by the addition ol 50 L of saturated sodium bicarbonate, and after 10 mm. diluted by 100 mL of diethyl ether. The organic portion was then washed by (2 x 50 mL) of both saturated sodium bicarbonate and brine. The combined organic layers were then dried over magnesium suLfate, filtered and concentrated vacuo. The crude oil was purified by silica gel chromatography elutmg with 30 ethyl acetate: hexane to provide 8.8 g of product (2 mmol, 75%) .
Figure imgf000137_0001
Figure imgf000137_0002
Boc am e
HCl/dioxane deprotected dιamme-2HCl
Figure imgf000137_0003
carbonyl diimidazole 162.15g/Mol 2.77g 17.1 mmol triethylamine 12.7mL 179 mmol methylene chloride 550mL 0.03 M
The Boc amme of Example IB was stirred in 15 mL of 4N HCl at 25 °C for 1.5 hours. The reaction mixture was then concentrated in vacuo to provide a white foaming solid. 3.83 mg of the deprotected diamine was dissolved m 500 L of methylene chloride. To this, triethyl amme was added. After stirring for 20 minutes, CDI was added (solid) . The reaction was then stirred for 24 hours. This was followed by concentration vacuo. The crude material was purified by silica gel chromatography, eluting with ethyl acetate, to provide 2.15 g (67 %) of the desired allyl urea. E am le 2
A .
Ph toluene.CH2CI2 ° Ph
Ptf cΛ ° *
Figure imgf000138_0001
2 3 Ptf < ~ ,OMe H u
1 aldehyde 1.0 equiv., 2 methyl 1.05 eq. (triphenylphosphoranyli dene)acetate
3 toluene 80mL
4 methylene chloride 120mL
Combine 7.9g of (S)-N-Boc-amino-3-phenyl-l-propanal, 40mL of anhydrous toluene and 60mL of anhydrous methylene chloride. Add 9.8g of the ylide followed by 20mL of toluene and 60mL of methylene chloride. Stir overnight at room temperature. After approximatly 18 hours the solvent was removed m vacuo and the residue was purified by flash chromatography (EtOAc/Hexane) to give 7.1g(77%) of the desired ester.
B .
Figure imgf000138_0002
1 1 ester 4.5g, 1.0 equiv,
2 magnesium turnings (Aldrich) 3.2g 10.0 eq..
3 2N HCl @ 10 eq.
To a solution of ester 1 in anhydrous methanol at 0 °C was added Mg turnings with stirring under N2. Bubbling became evident within 1 hour. The reaction was then stirred at 0 °C for -2.5 hours then allowed to warm to RT overnight (TLC (95:5, CH2Cl2:MeOH) showed reaction complete, st. mat. Rf = .84, prod. Rf = .25) . The reaction was cooled to 0 °C, neutralized with 2 N_ HCl, diluted with water, and the volume reduced in vacuo . The remaining aqueous layer was extracted with 3 portions of methylene chloride and the combined organic layers were washed with brine, dried (MgSU ), filtered, and concentrated in vacuo .. The residue was then purified by silica get flash chromatography (CH2Cl2 -- >3% MeOH/CH2Cl2) to yield desired lactam product (1.74g, 75% yield) . Literature reference: Tetrahedron . Let t . , 1993, 34 (28), pp. 4439-4442.
Figure imgf000139_0001
1 lactam from 2B 1.0 equiv., 1.7g
2 BOC anhydride 2.5 equiv., 5.2g
3 triethylamine 2.0 equiv, 2.7mL
4 DMAP 1.2 equiv, 1.4g
Lactam 1 was dissolved in methylene chloride (20mL) and to this solution was added a solution of Boc anhydride 2 in CH2C12 (10 ml) followed by triethylamine (2 eq) and DMAP (1.2 eq) . After stirring for 4 hours at room temperature the reaction was refluxed for 4 hours and after this time, an additional l.Og of Boc anhycride acetonitrile (20mL) and 700uL of triethylamine were added. The reaction was stirred for 15 hours at room temperature. (TLC (95:5, CH2C12 : MeOH) Rf (st mat.) = .31. Rf(prod) = .66.) The solvent was then removed in vacuo and the residue was partitioned between methylene chloride and water. The organic layer was washed with water and brine, dried (MgS0 ) and filtered. The dried organic layer was then concentrated in vacuo and the residue was purified by silica gel chro atagraphy (CH2C12) to yield desired boc lactam 2 {2.3g, 86%) .
D,
Figure imgf000140_0001
1 BOC-lacta from 2C 1.0 "equiv. , 85 mg
2 Allyl Bromide, (Aldrich) 1.8 equiv. , 51uL
3 LDA, 1.29M (Aldrich) 2.0 equiv , 420 uL
Boc-lactam 1 was dissolved in dry THF and cooled to -78 °C and to this solution was added LDA via syringe. After stirring for 40 mm. at -78 °C, allyl bromide was added via syringe and the reaction was stirred for 3 hours after which time an additional amount of allyl bromide (17 ul) was added. The reaction was then stirred at -78 °C for 4 hours (TLC (5:95, MeOH:CH2Cl2) Rf (st mat.) = .34. Rf(2 diast.) = .55 and.61) . The reaction was then quenched with lmL saturated NaCl solution, and partitioned between saturated sodium bicarbonate and ethyl acetate. The organic layer was then washed with water and brine, dried (MgS04), filtered and concentrated in vacuo . The residue was purified by silica get chromatography to yield allylated product 2 (47mg, 48% yield) .
E.
Figure imgf000141_0001
A mixture of diisopropylamme (4.6 L, 3 eq) and THF
(10 mL) was cooled to -78 °C, and to this solution was added n-butyl lithium (1.4 eq) via syringe. This mixture was warmed to -10 °C and stirred for 40 mm, after which time the mixture was cooled back to -76 °C. A solution of Boc lactam 1 (3.0 g, 1 eq) in THF (15 mL total) was added. The reaction mixture was then stirred at -78 °C for 40 min followed by the addition of benzyl bromide (1..45 mL, 1.1 eq) via syringe . After stirring for 2.5 hours at -78 °C, the reaction was warmed to -45 °C and stirred an additional 1 hour. The reaction was then quenched at -78 °C, with 0.5 mL saturated NaCl solution. The reaction was warmed to room temperature, diluted with ethyl acetate and the organic layer was washed with water and satuated NaCl, dried (MgS04) and concentrated in vacuo. The residue was then dissolved in methylene chloride (50 mL) and to this solution was added triflouroacetic acid (8 mL, excess) . After 4 hours the reaction was concentrated in vacuo, and partitioned between a saturated solution of sodium bicarbonate and ethyl acetate. The organic layer was washed with water and brine and then dried (MgSθ4) and concentrated in vacuo. The resulting residue was purified by flash silica get chromatography to give 726 mg (30%) of the desired benzyl lactam product 2 as a mixture of diastereomers.
Example 3 A.
Figure imgf000142_0001
Synthesis of 2-oxo-3-methyl-6-phenvlmethvlmorpholme. Dissolve S- (-)-2-Ammo-3-phenyl-l-propanol (1.51 g, 10 mmol) m THF (10 ml) . To 0 °C solution add (rac)-2— bromopropionyl bromide (1.04 ml, 10 mmol), followed by a dropwise addition of diisopropylethylamme (1.73 ml, 10 mmol) . Warm up to rt and continue stirring for 90 mm. Remove solvents m vacuo and remove salts by ethyl acetate/water extraction (3X) . Following magnesium sulfate drying, the ethyl acetate layer is evaporated and residue redissolved in anhydrous THF. To 0 CC solution of intermediate 2 add 13 mM of NaH (from 60% mineral oil dispersion, removed by washing, with hexane) . Solution was warmed up to rt and reaction terminated (MeOH) after 1 hr. Residue left after solvents removal was again partitioned between ethyl acetate/water (2X) , organic phases combined, dried with magnesium sulfate, filtered and evaporated, resulting in 1.20 g crude product. Silica gel chromatography (ethyl acetate) yielded 0.70 g of pure product, 34% yield. 1H NMR (CDC13): 7.25 (m, 5H) ,
6.75 (broad s, IH) , 4.19 (q, IH, J=7.0 Hz), 3.76 (2H, d, J=7.5 Hz), 3.57 (IH, m) , 2.90 (2H, m) , 1.49+1.46 (both s, total integration 3H) . CHN: 70.0 (calc: 70.2), 7.3 (7.4), 6.8 (6.8). Mass Spec. (API-)=204 (M- 1). Silica gel plates: Rf=0.19 (1/1 ethyl acetate/hexane) . HPLC at 220 nm (YMC 0.46 cm x 25 cm C18 reverse phase) t=11.47 mm (single peak), gradient: 0-100%B/30 mm, 1.5 ml/mm, A=O.I% TFA m water, B=0.1% TFA in acetonitrile. B .
Figure imgf000144_0001
Synthesis of 2-oxo-3, 3-dimethvl6- phenylmethvlmorpholine.
Dissolve 3.02g (20 mM) of S- (-)-2-Amino-3-phenyl-l- propanol in 10 ml THF. To 0 °C solution add 2-
Bromoisobutyryl bromide (2.47 ml, 20 mmol), followed by dropwise addition of diisopropylethylamme (3.47 ml, 20 mmol) . Warm up to rt and continue stirring for 90 mm. Remove solvents in vacuo and remove salts by ethyl acetate/water extraction (3X) . Following magnesium sulfate drying, the ethyl acetate layer is evaporated and residue redissolved in anhydrous THF. Following silica gel chromatography (1/1 ethyl acetate/hexane), 1.20 g of intermediate 2 is isolated from mixture containing overacylation product.
To 0 °C solution of 2 in 4 ml of anhydrous DMF add 4 mM of NaH (from 60% mineral oil dispersion, removed by washing with hexane) .
After 14 hrs at rt, the solvent was removed and solid residue partitioned between ethyl acetate/water (2X) , organic phases combined, filtered, evaporated and (silica gel) chromatographed with ethyl acetate, resulting in 0.20 g of product homogenous by TLC, but heterogeneous by HPLC.
C.
Figure imgf000145_0001
Synthesis of 2-oxo-3.3-spιrocy lohexvl6- phenylmethylmorpholme via multiple deprotonation- alkylation route.
A solution of 1 (5.73 g) was dissolved in 5 ml of anhydrous DMF, cooled down to 0 C and 0.72 g of NaH was added portionwise. After stirring for 15 min at room temperature, the solution was cooled to 0 C and 4.70 g of p-methoxy-benzyl chloride was added. The reaction was then stirred at room temperature for two hours, followed by silica gel purification, yielding 4.72 g (51%) of 2.
M (AP+) =312.1 (M+l) . 1H NMR (CDC13) =7.26-6.87 (9H,m), 5.42 (lH,d), 3.85 (lH,d), 4.34 (lH,d), 4.20 (d, IH) , 3.79 (s,3H), 3.68 (lH,d), 3.42 (lH,d), 3.26 (lH,m), 2.95 (2H, m) .
4.70 g of 2 was dissolved m 10 ml of anhydrous THF, cooled to -78 °C and 9.8 ml of 2M LDA in heptane/THF/ethylbenzene was added. After 15 mm, 4.56g of l-chloro-5-ιodopentane was added dropwise and the reaction carried out at -78 °C for 1 hr and then quenched. The solvents were removed and the material was purified by silica gel (2.6g, 41.4%) . The resulting compound (3) was ca 1:1 mixture of two diastereomers. MS (API+)=416.2 (M+l) . 1H NMR (CDC13)= 7.4-6.9 (9H, m) , 5.40 (IH), 4.23 (IH), 3.83 (IH), 3.80 (s,3H), 3.75 (IH), 3.55 (3H), 3.36 (IH), 3.12 (IH), 2.96 (IH), 1.88 (m,4H) , 1.58 (m,4H) .
2.6 g of 3 was dissolved in 5 ml of acetone. 1.87 g of sodium iodide was added and refluxed overnight. Acetone was then removed in vacuo and the crude material purified by ethyl acetate/aqueous extraction, resulting in 2.8g of 4 (88.3%) . MS (API+)=508.1 (M+l), 530.1 (M+Na) .
2.8g of 4 was dissolved in 40 ml of anhydrous THF, cooled down to -78 °C, and 3.6 ml of 2M LDA was added. The reaction was allowed to progress for 2 hrs, with gradual temperature increase to room temperature. The residue was quenched with water, THF was evaporated and the crude material desalted between ethyl acetate/water, resulting in 1.90 g of 5. IH NMR (CDCl3)=7.35-6.83 (m, 9H) , 5.35 (d, IH) , 3.79 (s,3H), 3.76 (d,lH), 3.55 (m,2H), 3.23 (m, IH) , 3.0 (m,2H) , 2.0-1.05 (m, 10H) .
1.90g of 5 was deprotected by 9.61 g of CAN in 3/1
(v/v) acetonitrile/water overnight at room temperature.
The product 6 (0.50g) was purified on silica using EtOAc/hexane/methanol gradient. M (AP+) =259 (M+l) . IH NMR (CDC13)=7.22 ( , 5H) , 6.96 (s,lH), 3.82 (m, IH) , 3.67 (m, IH) , 3.60 ( , IH) , 2.83 (m,2H) , 2.0-1.20 (m, 10H) .
Example
Figure imgf000147_0001
7.0g of the aldehyde 1 was dissolved in 40 mL of THF and added dropwise to a cooled (-78°) solution of 128 L (128mMol) of 1M trimethylsilyl methylmagnesium bromide in ether. The resulting mixture was allowed to warm to rt and poured into water. After diluting with ethyl acetate and IN HCl, the layers were separated and the organic layer was washed with 10% aqueous sodium bicarbonate. Drying over magnesium sulfate and removal of the solvent in vacuo gave a viscous oil, which was re-dissolved m 150 mL of dichloromethane and treated dropwise with 15.6 mL of borontnfluonde etherate. The resulting mixture was stirred for 5 days at rt and then quenched with 10% NaOH. The organic layer was dried and evaporated and the residue was chromatographed on silica gel (20% ethyl acetate/hexanes) to give 5.2g of a yellow solid. Recrystallization from hexane yielded 4.6g of the desired alkene as a white solid in three crops. B .
Figure imgf000148_0001
2
2.0g P.lmMol) of the alkene from the previous step were mixed with 10 L of carbon tetrachloride and 1.4 mL (20mMol) of thioacetic acid. A spatula tip cf AIBN was added and the mixture was irradiated in a quartz vessel at 254nm for 2h. The resulting mixture was diluted with dichloromethane and extracted with satd. aqueous sodium bicarbonate. Drying and removal of the solvent, followed by chromatography on silica gel (15% ethyl acetate/hexane) gave the desired thioacetate (2.0g) as a pale yellow liquid which solidified on standing.
Figure imgf000148_0002
A solution of 0.85g of the thioacetate from the previous step in 30 mL of acetic acid and 15 mL of IN HCl was cooled on ice and exposed to a stream of chlorine gas for 2h. Ethyl acetate was added and the organic layer was separated, dried and co-evaporated with toluene to give the desired sulfonyl chloride as a white solid (1.05g).
0.7g of the sulfonyl chloride 2 obtained in the previous step were dissolved in 30 mL of 30% HBr in acetic acid. After 2h, the volatiles were removed in vacuo, the gummy residue was redissolved in lOOmL of chloroform and the solution was treated with l L of triethylamine. The mixture was stirred for lh and then extracted with IN HCl and 10% aqueous sodium bicarbonate. Drying over magnesium sulfate and removal of the solvent gave a brown oil which was chromatographed on silica gel (2% MeOH/dichloromethane) to give the desired sulfonamide as an off-white solid (0.305g). 1H-NMR (CDC13) : 2.20 (lH,m) , 2.48 (lH,m), 2.89 (2H,m), 3.10 (lH,m), 3.23 lH,m), 3.84 (IH, m) , 4.18 (IH, bs), 7.30 (5H,m). 13C-NMR (CDC13): 28.8, 42.0, 47.8, 56.2, 127.8, 129.1, 129.3, 136.6.
Example 5 Synthesis of Sulfamate A.
1 ) methylamine, EDCI, DMF
2) H2/ 5%Pd-C/methanol
Figure imgf000149_0002
Figure imgf000149_0001
A solution of 30g of Cbz- (L) -phenylalanine, 6.8g of methylamine hydrochloride, 14.8g of hydroxybenzotriazole and 22 L of N-methylmorpholine in 300 mL of dimethylformamide was cooled on an ice-bath and treated with 19.2g of EDCI. The mixture was allowed to reach rt overnight and then poured into 2000 L of water. The product was collected by filtration, dried and redissolved in 500mL of methanol and 300 mL of THF. Ig of 5% palladium on carbon was added and the mixture was stirred under hydrogen for 36h. Filtration and removal of the solvent, followed by short plug filtration through silica gel (5% MeOH(2M NH3)/ dichloromethane) gave the desire amine as a pale yellow solid (17g) .
B .
Figure imgf000150_0001
A solution of 1.22g (56 mMol) of lithiumborohydride in 28 mL of THF was treated with 14.2 mL (112mMol) of chlorotrimethyl silane. The resulting mixture was treated scoopwise with 5g (28mMol) of the amide from the previous step. After stirring at rt for 24h, 40 mL of methanol were added carefully, followed by 10 mL of acetic acid. Repeated evaporation from methanol gave a colorless glass, which was dissolved in lOOmL of 20% NaOH. Extraction with 4x50mL of chloroform, followed by drying and removal of the solvent gave a yellow oil which was chromatographed on silica gel (20% methanol (2M ammonia) /dichloromethane to give 1.5g of the desired diamine as a colorless oil, and 2.0g of recovered starting material.
0.15g of the diamine from the previous step were dissolved in 0.5 mL of pyridine and added dropwise to a refluxing solution of O.lg of sulfonyldiimide in 1.5mL of pyridine. Reflux was continued for 24h and the volatiles were removed in vacuo . The resulting brown oil was chromatographed on silica gel (20% methanol (2M ammonia) /dichloromethane) to give the desired sulfonylurea as a yellow oil (0.04g) . """H-NMR (CD30D) : 2.60 (3H,s), 2.86 (lH,dd), 2.96 (lH,dd), 3.15 (lH,dd),
3.47 (lH,dd), 4.18 (IH, m) , 7.22 (5H,m),
13 7 7..3388 ((llHH,,dd)) . 13C C--NNMMRR ((CCDD33OOED) : 31.8, 39.9, 50.0, 57.8, 126.5, 128.2, 129.0, 136.6
Example 6
Figure imgf000151_0001
Boc lactam 1 (1.27 g, leq) was dissolved in THF (27 mL) and cooled to -78 °C. To this solution was added LDA (Aldrich, 1.5 M in hexane, 3.7 mL, 1.2 eq) via syringe over 3 minutes. After stirring for 85 minutes at - 78 °C, a solution of ethyl iodoacetate (600 uL, 1.1 eq) in THF (13 mL) was added via syringe over 6 minutes. The reaction was then stirred at -78 °C for 4.5 hours, then at 1.5 hours at -40 CC. The reaction was then cooled back to -78 °C and quenched with 2.5 L saturated NaCl solution, and partitioned between saturated sodium bicarbonate and ethyl acetate. The organic layer was then washed with brine, dried (MgS04), filtered and concentrated in vacuo . The residue was purified by flash silica get chromatography eluting with 5% EtOAc/ CH2C12 to give 1.67g of substituted lactam product 2 contaminated with a minor amount of lactam starting material 1. HPLC showed 52% product and 28% starting material. This mixture was then dissolved in methylene chloride (45 mL) and cooled to 0 °C. To this solution was added trifluoroacetic acid (2 mL) and the reaction was stirred at room temperature for 1.5 hr. TLC showed no BOC material and the reaction was concentrate m vacuo and partitioned between saturated bicarbonate solution and ethyl acetate. The organic was washed with water, brine and dried (MgS0 ) . The organic layer was evaporated in vacuo, and the residue was purified by flash chromatography eluting with 3:1 EtOAc/ hexane to give 770mg of pure lactam product 2.
Example 7
Figure imgf000152_0001
A solution of 5-benzyl-pyrrolιdinone 1 (1.5 gr, 8.86 mmol) was dissolved at ambient temperature under nitrogen in anhydrous dichloromethane (40 mL) . TMEDA (6.5 mL, 42.8 mmol) was added via pipette and the solution was cooled and maintained at -20 °C. TMSI (2.33 mL, 17.12 mmol) was added via pipette and the mixture was stirred for 15 mm. Solid iodine (4.345 g, 17.12 mmol) was added and the mixture was stirred vigorously for 15 minutes and then quenched by rapid addition of the reaction mixture into aqueous 10- sodium sulfite solution (100 mL) . The mixture was transferred to a separatory funnel and the layers were separated. The organic layer was washed with IN aHSU , water, and then dried over MgS04. The solution was then diluted in half with methanol and stirred overnight under a nitrogen atmosphere. The solvent was removed in vacuo and the residue was purified by flash chromatography, elutmg with ethyl acetate : hexane (7:3) . Pure iodo lactam product 2 was recovered as a solid (2.11 g) .
Example 8
A.
Figure imgf000153_0001
To a solution of dibenzylphenylalmol 1 (100 mmol) in methylene chloride (lOO L) , was added triethylamine (150 mmol) . The mixture was cooled to 0 °C and methanesulfonyl chloride (110 mmol) was slowly added. The mixture was stirred at 0 CC for one hour and then poured into a beaker containing diethyl ether (400mL) . The mixture was filtered and washed with more diethyl ether and the filtrate was washed with water, saturated NaHC03 and saturated brine. The organic layer was then dried (MgS04), filtered and concentrated to yield 41 g of crude mesylate product 2 as a light yellow-brown thick oil, which was used as is m subsequent steps.
B.
Figure imgf000154_0001
Diethyl malonate (300 mmol) was dissolved in acetonitrile (250 mL) and to this solution was added potassium carbonate (300 mmol) ; the suspension was stirred overnight at room temperature. Mesylate 1 (100 mmol) m acetonitrile (60mL) was then added to the reaction mixture which was then heated to 80 °C and stirred overnight. The reaction mixture was then filtered and concentrated in vacuo. Addition of hexane to the residue formed a precipitate, which was filtered as pure malonate product 2 (19.5 g) . Material was used as is.
Figure imgf000155_0001
Malonate 1 (10.6 mmol) was dissolved in dry THF (40 mL) and cooled to 0 °C. To this solution, sodium hydride (17 mmol) was added in portions and the suspension was stirred for 1.5 hr at 0 °C. The triflate 2 (12 mmol) in dry THF (lOmL) was then slowly added to the reaction mixture and after complete addition the reaction was allowed to warm to room temperature and was stirred overnight. The reaction was then diluted with water (lOOmL) and extracted with diethyl ether (3x50 mL) . The combined organic layers were then washed with saturated brine, dried over MgS04, filtered and concentrated in vacuo. The crude product was purified by mplc (eluted with a gradient of 9:1 hexane:ethyl acetate up to 4:1 hexane:ethyl acetate to yield product 3 (4.2 g, 73 %) .
D.
Figure imgf000155_0002
2
The subsituted malonate 1 (1.62 mmol) was suspended m ethanol and to this was added cone. HCl (0.24 mL, 2.4 mmol) and 10% palladium on Carbon (0.162 mmol) . This mixture was then stirred under a balloon of hydrogen gas at room temperature overnight. The reaction was then filtered through Celite and to the filtrate was added triethylamine (10 L, excess) followed by solid sodium bicarbonate (excess) . The mixture was stirred for 0.5 hr, filtered and concentrated to yield a yellow solid. This residue was then dissolved m ethyl acetate and washed with water, 0.5N HCl, saturated sodium bicarbonate, and brine. The organic layer was dried (MgS04), filtered, and dried to yield crude lactam product 2, which was used as is.
Figure imgf000156_0001
Lactam 1 (1.18 mmol) was dissolved m ethanol (5mL) and to this solution was added KOH (10 mmol). The mixture was stirred for 3 hr at room temperature and then concentrated to dryness. The residue was dissolved m water and washed with diethyl ether. The aqueous layer was then acidified with HCl and extracted with ethyl acetate. The organic layer was dried (MgS04), filtered and concentrated in vacuo to yield 341 mg of a l ght yellow solid. The residue was dissolved in DMSO (3mL) and to this solution was added p-toluenesulfonic acid mono- hydrate, and the mixture was heated to 80 °C overnight. The mixture was diluted with water (15 L) and extracted with ethyl acetate. The organic layer was washed with saturated sodium carbonate and brine followed by drying with MgS04. The organic layer was then filtered and concentrated in vacuo to yield the THF substituted lactam product (245 mg, 77% from ester) which was used as is m the next step without further purification.
Example 9
A .
Figure imgf000157_0001
Sodium hydride (60% dispersion in mineral oil, 4.0 g, 1.17 eq) was washed with 4 x 25 L portions of hexanes to remove the mineral oil, then suspended in 25 L of DMF and cooled to 0 °C. A solution of lactam 1 (15g, 1 eq) dry DMF (25 mL) was then added dropwise via canula into the cold NaH suspension over 40 mm. An additional 65 mL of DMF was then added to aid stirring. After stirring the anion for 1 hour, p-methoxybenzyl chloride (14.5 L, 1.26 eq) was added over 5 mm at 0 °C. The reaction was then allowed to warm to room temp. An additional amount of p-methoxybenzyl chloride was added to drive the reaction to completion. TLC (EtOAC) Rf lactam 1 = 0.21. Rf product 2 = 0.43. After 3.5 hours, the reaction was poured into cold water and extracted twice with ethyl acetate. The combined organic layers were washed with water (5X) , brine, dried (MgS04) and filtered. Concentration in vacuo , afforded a crude solid which was purified by crystallization( 7:1 hexane:EtOAc) to yield the protected lactam product 2 (19g, 75%) .
B ,
Figure imgf000158_0001
To protected lactam 1 (328 mg, 1.11 mmol) and N,N,N ,N -tetramethylethylenediamme (Aldrich, 5.0 equiv., 5.55 mmol, 645 mg, 838 ml) m 15 ml dichloromethane at -15 CC, was added lodotrimethylsilane (Aldrich, 1.0 equiv., 1.11 mmol, 222 mg, 158 ml) . After 15 mm, iodine (Aldrich, 1.2 equiv., 1.33 mmol, 338 mg) was added in one portion and the reaction warmed to 0 °C. After 30 mm the reaction was quenched with 5 ml each of 10% aqueous sodium sulfite and saturated aqueous sodium chloride. The orgnic layer was separated, dried over magnesium sulfate, filtered and concentrated in vacuo .
Purification by flash column chromatography (silica gel, 2.5 x 10 cm, 2.5% diethylether in dichloromethane) yielded 322 mg of diastereomeric iodolacta 2 as a white solid.
C,
Figure imgf000159_0001
To iodolactam 1 (1.18g, 2.91 mmol) and methyl vinyl sulfone (Aldrich, 6.0 equiv., 17 mmol, 1.82 g, 1.5 ml) in 25 ml refluxing toluene was added tributyltin hydride (Aldrich, 1.3 equiv., 3.79 mmol, 1.10 g, 1.0 ml) and AIBN (Pfaltz & Bauer, 0.12 equiv., 0.35 mmol, 57 mg) as a solution in 5 ml toluene over 1.2 h. After 16 h the solvent was removed in vacuo, and the residue taken up in 200 ml diethyl ether and stirred with 20 ml 10% aqueous potassium fluoride (wt/v) at ambient temperature. After 3 h the orgnic layer was separated, dried over magnesium sulfate, filtered and concentrated in vacuo . Purification by flash column chromatography (silica gel, 5 x 20 cm, 2:1 ethyl acetate/hexanes) yielded 0.31g of diastereomeric sulfone 2 as a white solid. Exampl e 10
A .
HCl
Figure imgf000160_0001
To a solution of solution of Cbz-L-phenylalinal (13 g, 45.9 mmol) in 1%ACOH/DMF (200 mL) mL was added aminoisobutyic acid methyl ester hydrochloride 1 (8.5 g, 55.1 mmol) with stirring at room temperature. Once homogeneous, solid sodium cyanoborohydride (8.6 g, 137.6 mmol) was added in one portion. Some bubbling was evident and the reaction was stirred overnight at room temperature. The reaction was quenched with water (20 L) and concentrated in vacuo to about 100 L. The concentrate was diluted with ethyl acetate and washed with water and brine followed by drying ( gS04) . The organic layer was evaporated in vacuo to yield a yellow residue which was purified by MPLC (elutant 1:2 ethyl acetate : hexane) to afford amine product 2 (11.6 g, 66%) . B .
Figure imgf000161_0001
1
To a solution of amme 1 (1.41 gr, 3.7 mmol) in methylene chloride (25 mL) was added 30% HBr in acetic acid (6 mL) via pipet. Vigorous gas evolution occurred and the reaction was allowed to stir overnight at room temperature. The mixture was then evaporated m vacuo and dried under high vacuum. The residue was then dissolved m methanol (25 mL) and to this solution was added diisopropylethylamme (5eq) and the reaction was stirred at room temperature overnight. The solvent was removed in vacuo and the residue was taken up m ethyl acetate and washed with water, saturated NaHC03 and brine. The organic layer was dried (MgS04) filtered and concentrated in vacuo to yield crude product . Flash silica gel chromatography (8% methanol / methylene chloride) afforded pure piperazmone product 2 (556 mg, 70 %) .
Figure imgf000162_0001
To a solution of piperazinone 1 (556 mg, 2.55 mmol) and potassium carbonate (1.06 g, 7.6 mmol) in acetonitrile was added benzyl bromide (364 uL, 3 mmol) and the reaction was stirred at room temperature overnight. The reaction was then filtered and concentrated in vacuo. The residue was dissolved in ethyl acetate, washed with water and brine and dried (MgS04) . The organic layer was then removed in vacuo and the residue was flash chromatographed (3% methanol in methylene chloride) to yield pure benzyl protected piperazinone product 2 (589 mg, 75%) .
Example 11
A .
Figure imgf000162_0002
A solution of Cbz- (1) -Phenylalanine (15 gr, 50 mmol), HOBT (7.4g, 50mmol), N-methyl morpholine (5.5 mL,, 50 mmol) and benzylamine (6 mL, 55 mmol) in 250 mL of DMF was cooled to 0 °C and treated with EDCI (9.6 g, 50 mmol) . The resulting mixture was stirred at 25 °C for 12h and the volatiles were removed in vacuo. Partitioning between ethyl acetate and IN hydrochloric acid, followed by extraction with 10% sodium bicarbonate, drying over magnesium sulfate and evaporation of the solvent afforded the desired amide as a white solid (19.5g) . 19g of the above material were dissolved in 280mL of 30% hydrogen bromide acetic acid and stirred at 25 °C for 3h. The volatiles were removed and the residue was partitioned between water and ether. The aqueous layer was treated with excess 6N sodium hydroxide and extracted twice with ethyl acetate. Drying over magnesium sulfate and evaporation of the solvent afforded the desired amme as a pale yellow oil (14. Og), which was redissloved in 200 mL of tetrahydrofuran and treated with 200 mL of 1M borane- THF in tetrahydrofuran. The mixture was stirred at 25 °C for 72h and then heated to reflux for 4h. The solution was cooled and treated with 100 L of methanol under vigorous gas evolution. The volatiles were removed and the resulting residue was dissolved m 150 mL of concentrated hydrochloric acid. After refluxmg for lh, the volatiles were removed and the residue was dissolved in 300 mL of 3N sodium hydroxide. Extraction with 3 times 250 mL of dichloromethane, drying over magnesium sulfate and chromatography on 2 inches of silica gel (2% methanol-dichloromethane) gave the desired diamine as a pale yellow honey (9.2g) . B .
Figure imgf000164_0001
A solution of sulfonyldiimide (3.6 g, 36 mmol) in 100 mL of pyridine was heated to reflux and treated dropwise with a solution of the diamine 1 (7.2 g, 30 mmol) from the previous step in 20 mL of pyridine.
After 2h of reflux, 15 mL of triethylamine and 0.4g of 4-dimethylaminopyridine were added and heating was continued for 12h. The volatiles were evaporated and the residue was partitioned between IN hydrochloric acid and ethyl acetate. Extraction of the organic layer with saturated sodium bicarbonate, drying over magnesium sulfate and chromatography on silica gel (1:1 ethylacetate - hexanes) afforded the desired cyclic sulfamate 2 as a white solid (6.0g) . 1H-HMR (CDC13) : 2.80 (IH,dd) , 2.96(lH,dd), 2.98(lK,dd), 3.32(lH,dd) , 3.95(lH,m), 4.04(lH,d), 4.24(lH,d) , 4.40(lH,d) , 7.18(2H,d), 7.2-7.4(8H)
13C-NMR(CDC13) : 41.5, 50.0, 52.7, 53.8, 127.5, 128.0, 128.2, 128.3, 28.4, 128.5, 135.5, 136.0 Example 12
Figure imgf000165_0001
The Cbz-phenylalaninol mesylate 1 (280 mg, 0.77 mmol) was stirred in acetonitrile (5 L) containing benzyl amine (413 mg, 3.85 mmol) and sodium iodide (115 mg, 0.77 mmol). The reaction was then refluxed for 24 hours. The reaction was then cooled to 25 °C and concentrated in vacuo. The crude oil was then purified by silica gel chromatography, eluting with CH2C12 with a gradient up to 1:1 CH2Cl2:EtOAc to provide 120 mg of the desired diamine 2.
B.
Figure imgf000165_0002
1
The Cbz protected diamine 1 (120 mg, 0.32 mmol) was stirred in 2.0 mL of 30 % HBr in acetic acid for one hour. This was followed by concentration in vacuo, The crude oil was then dissolved into toluene and concentrated in vacuo two times followed by evacuation at approx. 1 mm Hg. The crude diamine was then purified by silica gel chromatography, eluting with 95:5:1, CH2C12:MeOH:NH4OH to provide 71 g ( 90 %) of the desired diamine 2.
Figure imgf000166_0001
The diamine 1 (56 g, 0.23 mmol) was dissolved in 3.0 mL of CH2C12. This was followed by the addition of TEA (66 uL, 0.25 mmol) and then CDI (32 mg, 0.25 mmol) . A new spot was observed by tic after 2-3 hours (Rf = 0.29 in EtOAc on Si02) . The reaction mixture was then concentrated and the residue was purified by silica gel chromatography, eluting with EtOAc, to provide 32 mg (52%) of the desired benzyl urea 2.
Example 13
Synthesis of Compound 1
Figure imgf000167_0001
al lyl urea 216 g/Mol lOOmg 0.46 mmol
NaH, ( 60% in oil ; 24 g/Mol 140.0 mg 9.7 mmol epoxide 325.4 g/Mol 150.0 mg 0.46 mmo1
DMF 2.0 mL
The urea of Example IC was dissolved in 1.0 L of anhydrous DMF and cooled to 0 °C. This was followed by the addition of 140 mg NaH. The reaction turned darker over the next hour at 0 °C. This was followed by the dropwise addition of the epoxide as a solution DMF (0.6 mL) , washing with 300 uL of DMF. The reaction was then stirred one hour at 0 °C, followed by warming to 25 °c. Tic indicated nearly complete conversion to two new products (Rf = 0.4 and 0.45 on Si02 with 2:1 hexane: ethyl acetate, between that of the epoxide and the urea) . The reaction was then cooled to 25 °C and quenched by the addition of 3 mL of saturated sodium bicarbonate. The reaction mixture was then diluted by 15 L of methylene chloride and washed by both saturated sodium bicarbonate and brine, (2 x 15 mL each) . The organic portions were then dried over sodium sulfate, filtered and concentrated in vacuo . The crude product was then purified by silica gel chromatography, eluting with 80% ethyl acetate : hexane to provide 35. 0 mg of the desired alcohol .
Example 14
Figure imgf000168_0001
1 lactam 1.0 equiv., 295τιgg
2 sulfonamide epoxide 1.1 equiv., 520mg
3 NaH, 60% in oil (Aldrich) 1.5 equiv, 102mg 4 DMF 8 mL
Lactam 1 was dissolved in 3mL of DMF and cooled to 0 C. To this solution was then added sodium hydride as a solid and the reaction was stirred for 40 mm. at 0 °C. The anion solution was canulated into a solution of epoxide 2 in 3 mL of DMF. The reaction was stirred at 0 °C for 5 minutes, then warm to room temperature and stirred overnight (TLC (95:5, CH2C12 : MeOH) Rf (st mat.) = .26. Rf (prod) = .46) . After 22 hours, the reaction was cooled to 0 °C, and quenched with H20/EtOAc. The organic layer was washed with water (5X) and brme, dried (MgSθ4), filtered, and concentrated m vacuo . The residue was then purified by silica gel chromatography (40% ether/ CH2C12) to yield product 3 (310mg,37%) .
Example 15
A.
Figure imgf000169_0001
lactam 1.15g, 1.0 equiv. t-butyldimethylsilyl 1.5 equiv. + .5 eq. , trifluoromethanesulfonate (1.06mL) imidazole 2.5 equiv + .5 eq, (470mg!
Lactam 1 was dissolved 5mL of DMF and cooled to 0 °C. To this solution was then added imidazole followed by TBDMS-triflate. The reaction was then allowed to warm to room temperature. After approximatly 2 hours, an additional .5 eq. (80mg) of TBDMS-triflate and .5 eq. (265uL) of imidazole was added and the reaction was stirred overnight. The reaction was quenched with saturated NaHC03 solution and partitioned between H20/EtOAc. The organic layer was washed with water(5X) and brme, αried (MgSθ ), filtered, and concentrated in vacuo to yield product 2 (1.5 gr,37%) which was used as is.
Example 16 Synthesis of Compound 7
Figure imgf000170_0001
silyl-lactam 1.0 equiv., 23mg
Allyl Bromide, (Aldrich) 2.1 equiv. , 7 uL
LDA, 1.29M (Aldrich) 1.25 equiv , 36uL
TBAF, 1.0M, (Aldrich) 2.5 equiv., 95uL:
Silyl protected lactam 1 was dissolved in THF and cooled to -78 °C. To this solution, was added LDA (1.25 eq) via syringe. After stirring for 30 minutes at -78 °C, allyl bromide was added via syringe. After 2 hours an additional 2ul of allyl bromide was added and the reaction was stirred at -78 °C for 2.5 hours, then warmed to room temp for 17 hours (TLC (2:8, ether:CH2Cl2) Rf (st mat.) = .56. Rf (silyl-prod) = .72) . After this time, TBAF (1M in THF) was added and the reaction was stirred at room temperature for 7 hours (TLC (1:9, ether:CH2Cl2 ) Rf(prod) = .20) . The reaction mixture was then partitioned between H20/EtOAc and the organic layer was washed with water and brme, dried (MgS04) and filtered concentrated in vacuo . The residue was then purified by silica gel chromatography (10% ether/methylene chloride) to yield product 2 (6mg, 30% yield) .
Example 17 .Synthesis of Compound 20
Figure imgf000171_0001
silyl-lactam 1.0 equiv., 122mg benzyl bromide, (Aldrich) 1.5 equiv. 42uL
LDA, 1.29M (Aldrich) 1.4 equiv , 275ul
TBAF, 1.0M, (Aldrich) 2.5 equiv., 625uL
Silyl lactam 1 was dissolved in dry THF (6mL) and cooled to -78 °C. To this solution was then added LDA and the reaction was stirred for 30 minutes at -78 °C after which time benzyl bromide was added via syringe. The reaction was stirred at -78°C until reaction was complete (1.5 hours, TLC (1:9, ether:CH2Cl2 ) Rf (st mat.) = .29. Rf (silyl-prod) = .62. Rf (BzBr) = .79) .
The reaction was then quenched at -78 °C with 6uL water and then TBAF (IM in THF was added and the reaction was warmed to room temperature and stirred for 3 hours (TLC (1:9, ether:CH2Cl2 ) Rf (prod) = .28). The reaction was partition betweem H20/EtOAc and the organic layer was washed with with water and brine, drigd (MgS04) and filtered and concentrated in vacuo . The residue was purified by silica gel chromatography (10% ether/ CH2C12) to yield benzyl product 2 (71mg, 48% ) .
Example 18
Synthesis of Compound 1 6
Figure imgf000172_0001
silyl-lactam 1.0 equiv., 66mg
Methyl iodide, (Aldrich) 1.6 equiv., 16ul
LDA, 1.29M (Aldrich) 1.3 equiv , HOuL
TBAF, 1.0M, (Aldrich) 3.0 equiv., 325uL:
The reaction for the above methylated compound was carried out as per the procedure described for compound 20 (Example 17) substituting methyl iodide for benzyl bromide on the scale described in the above table. The final compound was purified by silica gel chromatography using 10% ether/ CH C12 to yield methylated product 2 (33mg, 60% yield) . Example 19
Figure imgf000173_0001
Figure imgf000173_0002
Lactam 1 was dissolved in dry DMF (15 mL) and cooled to 0 °C under a nitrogen atmosphere. To this solution was added sodium hydride (2 eq) m one portion and the reaction was stirred at 0 °C for 1 hour after which, epibromohydrm was adαed via syringe. After stirring for 5 mm. at 0 °C the reaction was warmed to room temperature (TLC (EtOAc) Rf (st mat.) = .16.
Rf(prod) = .23) . After 1.5 hours at room temperature the reaction was quenched with saturated NH4CI and extracted with CH C12. The organic layer was then washed with water (4X) and brme, dried (MgS04) and filtered, and concentrate in vacuo . The residue was then purified by silica gel chromatagraphy (3:1 EtOAC:hexane) to yield 315mg(60%) of epoxide product 2 which was used as is in the next step. B ,
Figure imgf000174_0001
lactam 1.0 equiv., 315mg cyclopentylmethylamine 5.75 equiv., 775mg anhy. EtOH 3mL
Epoxide 1 was dissolved in 3 mL of EtOH and to this solution was added cylcopentylmethyla ine. The reaction was heated to 80 °C for 2.5 hours (TLC
(9:l,CH2Cl2:MeOH) Rf (st mat.) = .56. Rf (prod) = .13) . The solvent was removed in vacuo and the residue was purified by silica gel chromatagraphy (3%MeOH/ CH2C12 to 10%MeOH/ CH2C12) to yield 224mg(50%) of amme product 2.
C. Synthesis of Compound 15
Figure imgf000174_0002
1 lactam 1.0 equiv., 315mg chlorotπmethylsilane 2.2 equiv., 112uL triethylamine 5.0 equiv., 280uL
4-metnoxybenzenesulfonylchloπde 1.5 equiv., 124 mg
TBAF, 1.0M 4.4 equiv., 1.78mL
Amme 1 from Example 19B was dissolved in methylene chloride and cooled to 0 CC. To this solution was added triethylamine (2.5 eq) followed by chlorotπmethylsilane. The reaction was then warmeα to room temperature and stirred under nitrogen for 2.0 "hours. An additional amount of triethylamine was added (2.5 eq) and 4-methoxybenzenesulfonyl chloride was added. The reaction was stirred at room temperature for 3 hours. After this time, TBAF (IM in THF) was added and the reaction stirred at room temperature for 1 hour. The solvent was removed m vacuo . and the residue partitioned between ethyl acetate and aqueous saturated bicarbonate solution. The organic layer was washed with water, br e, dried MgSθ , filtered and tne solvent removed m vacuo . (TLC (8:2, CH2C1 : ether), Rf (upper diast.) = .21 Rf (lower diast.) = .12) . The residue was purified by silica gel chromatagraphy (251 ether/CH2Cl2) to yield 52 mg(26%) of (upper diastereomer) . The lower diastereomer was further purified by preperative TLC (1:1, ether:CH2C12) to give 23mg(12%) of tne lower diastereomer. Example 20
Synthesis of compound 47
Figure imgf000176_0001
Morpholinone 1 was dissolved in 1 ml of anhydrous DMF, cooled to 0 C and to this solution was added 4.4 mg of NaH. The solution was brought to room temperature for 30 min and then cooled down to OC before adding 0.20 g of epoxide 2. After heating for 5 hrs at 45 °C, the solvent was removed in vacuo and purified on silica gel yielding 111 mg of final product 2 (compound 47) . M (ES+) =585 (M+l), 607.1 (M+Na) . IH NMR (CDC13)= 7.52 (d, 2H) , 7.30 ( , 5H) , 6.95 (d, 2H) , 4.05 (m, IH) , 3.87 (3H, s), 3.60 (m, 2H) , 3.16 (m, 4H) , 3.0 (m, 4H) , 2.18 (IH, m) , 1.97 (m, 2H) , 1.60 (m, 14H), 1.23 (m, 4H) .
Example 21 Synthesis of Compound 109
Figure imgf000177_0001
To a cooled solution (-78 °C) of benzyl lactam 1 (0.150g, 0.57 mmol) and bromomethyl acrylic acid (0.094g, 0.57mmol) in anhydrous THF (4.0mL) was added NaH (60%, 0.046g, 1.14 mmol) with stirring. The solution was allowed to gradually warm to room temperature and stir for 1.5h. The reaction mixture was then diluted with ethyl acetate (60mL) and washed with 1.0N HCl (2 x lOmL) and brme (2 xlOmL) . The organic layer was dried (magnesium sulfate) , filtered, and evaporated to give an off white solid. This solid was dissolved m methylene chlonde/methanol (80/20, lOmL) and through the cooled solution(-78 CC) was bubbled ozone for 10mm. The solution was flushed with oxygen, warmed to 0 °C, and methyl sulfide (2.0mL) was added at 0 °C. The mixture was allowed to warm to room temperature and stand for l.Oh. Evaporation of the solvent afforded crude product 2 as a yellow oil. To a solution of the acid 2 m anhydrous DMF (3.0mL) was added thioprolme-t-butylamide (O.llg, 0.57mmol), hydroxybenzotriazole (0.77g, 0.57 mmol), N- metnyl- morpholine (0.62mL, 0.57mmol) and EDCI (O.llg, 0.57 mmol) respectively with stirring at room temperature. After 24h. at room temperature, the reaction mixture was evaporated and the residue was dissolved in ethyl acetate (lOOmL) . The solution was washed with 1.. ON HCl (2 x 20mL) , 10% sodium carbonate (2 x 20mL) , water (1 x lOmL) , brine ( 1 x lOmL) , filtered and evaporated to give 0.210g of a yellow oil. The oil was purified by column chromatography; hexane/ethyl acetate (60/40) to give compound 3_ (0.050g, 18%) MS: M+l= 522; H NMR (chloroform-d) 1.35 (d, 9H) ; 1.85 ( , 2H) ; 2.6 (m, 3H) ; '2.85(m, IH) ; 3.15(m, 2H) ; 3.40 (m, IH) ; 3.8(m,lH) ; 4.1 (m, 2H) ; 4.4(m,lH); 4.70(m,lH) ; 4.95(m, IH) ; 6.1(d, IH) ; 7.1 (m,4H) ; 7.25 (m,6H) .
Example 22
Synthesis of Compound 80
Figure imgf000178_0001
0.80g of allyl lactam 1 was dissolved in 1 ml of DMF, cooled to 0 °C and 89.5 mg of sodium hydride was then added. The solution was then brought up to ambient temperature for 30 min, again cooled down to 0 °C and 1.4 g of epoxide 2 was added. The reaction was warmed to 50 °C under N2 blanket for 3 hrs. The resulting crude mixture was then chromatographed on silica gel yielding 1.4g of 3 (63.7%). This amount was treated with 12 ml of 4N HCl in dioxane and 2 ml water for 30 min. The product was then chromatographed on C18rphplc, yielding 0.36g of two diastereomers, subjected to chiral separation, which resulted in 138 mg of pure diastereomer 3. MS (ES- 551.3 (M-l)), ES+, 553.3 (M+l) and 575.3 (M+Na) . IH NMR (CDC13)= 7.20 (m, 14H) , 6.26 (m, IH) , 5.62 ( , IH) , 5.24 (m, IH) , 4.97 (m, 2H) , 4.23 (m, IH) , 3.83 (m. 2H) , 3.61 (m, IH) , 2.95 (m, 10H) , 2.40 (m, IH), 2.24 (m, IH) , 2.04 (m, IH) , 1.95 (m, 2H) , 1.70 (m, 2H) .
Example 23 Synthesis of Compound 91
Figure imgf000179_0001
A solution of cyclic sulfamate 1 (O.lg, 0.33mmol) 2 mL of dimethyl formamide was cooled to 0 °C and treated with of 60% sodium hydride (0.005g, 0.13 mmol) in oil. The mixture was stirred at 25 CC for 1.5h and treated with of epoxide 2 (0.125g, 0.33mmol) The resulting mixture was stirred at 60 °C for 3h, more sodium hydride (0.005g) was added and heating was continued over night. The volatiles were removed m vacuo and the residue was dissolved in 2 L of 4M hydrogen chloride in 1,4-dιoxane. Water (0.5 mL) was addec and tne mixture was stirred for 6h at 25 °C. The reaction- mixture was diluted with ethyl acetate and extracted with 10% sodui bicarbonate. Drying over magnesium sulfate and removal of the solvents gave a yellow gum, which was subjected to C-18 preparative HPLC (acetonitrile-water gradient) . The desired material 3 was isolated as a minor fraction (9 mg) as a white solid 1H-NMR(CDC13) : 2.10(2H), 2.70(2H), 2.8-3.2(8H), 3.4(1H), 3.58(1H), 4.02(1H), 4.15(1H), 4.22(2H), 5.30(1H), 5.86(1H), 7.06(2H), 7.1-7.4 (16H) .
Example 24
Synthesis of Compound 83
Figure imgf000180_0001
To a cooled solution (0 °C) of compound 1 (0.190g, C.72mmol) in anhydrous DMF (lOmL) was added NaH (60%, 0.028g, 0.72mmol) with stirring. The solution was allowed to warm to room temperature and stir for l.Oh. Compound 2 (0.275g, 0.73mmol) was added at room temperature and the mixture was heated at 60 CC for 5.Oh. The solution was evaporated and the reside was partioned between ethyl acetate (150mL) and water (30mL) . The organic layer was washed with water (2 x lOmL) , brine (25mL) , dried (MgS04), filtered, and evaporated to give a grey oil. The oil was purified by column chromatography: hexane/ethyl acetate (60/40) to give 0.23g (50%) of the acetonide protected product. The acetonide (0.185g, 0.29mmol) was dissolved in isopropanol (lOmL) and treated with cone. HCl (3.0mL) at room temperature. After 1.5h., the solution was adjusted to pH 11 with 3.ON NaOH and then concentrated. The aqueous solution was extracted with ethyl acetate (3 x75mL) . The ethyl acetate was dried (MgSOή) and evaporatated to give a clear film. The crude product was purified by column chromatography: hexane/ethyl acetate (45/55) to give the product as a white solid (0.090g, 50%). Preparative HPLC on chiral phase (isopropanol-hexane gradient) yielded the desired diastereomer 3 (lOmg) along with a 1:1 mixture of the desired diastereomer and an additional epimer (50mg) . MS: M+l= 603 H NMR (chloroform-d) 1.80(m, 6H) ; 2.50 (m, IH); 2.60 (m, 2H) ; 3.0(m,8H); 3.60 ( , IH) ; 3.70(m,lH); 3.95(m,lH); 4.25(m,lH); 5.30(m,lH); 6.00(m,lH); 7.05(m,4H); 7.25(m,15H).
Example 25
Synthesis of Compound 8 A.
Figure imgf000182_0001
Allyl lactam 1 (443 mg, 2.06 mmol) was dissolved in DMF (2 L) and to this solution was added sodium hydride (2.2 mmol) . The reaction mixture was stirred at room temperature for 1 hr after which (s) -epichlorohydrin (172 ul, 2.2 mmol) was added neat. The reaction was stirred at room temperature for 4 hr, diluted with water (20 mL) and extracted with ethyl acetate. The organic layer was then washed with water, brine and dried (MgSθ ) and filtered. Concentration in vacuo afforded crude epoxide product 2 which was used without further purification.
B.
Figure imgf000182_0002
Lactam epoxide 1 (180 mg, 0.66 mmol) and decahydroisoquinoline 2 (160 mg, 0.66 mmol) were heated to 80 °C in isopropanol. After three hours the reaction was cooled to 25 °C and stirred for 48 hours at room temperature. The reaction was then concentrated m vacuo . Purified by silica gel cnromatography, eluting with 25 % EtOAc : Hexanes, providing 90 mg (90% pure by HPLC) of desired product 3.
Example 26 Synthesis of Compound 9 A.
Figure imgf000183_0001
Tne Boc protected piperazine 1 (21.4 mg, 0.081 mmol), was dissloved in 1.5 mL of 1-PrOH. This was followed by the addition of the lactam epoxide 2 (18.3 mg, 0.068 mmol) . The reaction vessel was then fitted with a reflux condenser and heated to 75 °C for 16 hours. TIC indicated complete consumption of both starting materials and formation of a new material. The reaction was then cooled to 25 °C and concentrated in vacuo. The complete consumption of epoxide was confirmed by both tic and H NMR. The crude additic product was then used without further purification. B .
Figure imgf000184_0001
The Boc protected piperazine addition product 1 from the previous step was stirred for 2 hours in 1.0 mL of 4N HCl/dioxane. This was followed by concentration vacuo. The crude solid was then dissolved in 10 mL of
CH2C12 and washed by 2 x 10 mL of each saturated aqueous sodium bicarbonate and saturated aqueous brme. The combined organic portions were then dried over MgSOή, filtred and concentrated m vacuo to provide the freebase of the desired intermediate. The crude amme was then dissolved in 1.0 mL of DMF at 25 °C. This was followed by the addition of the hydrochloride salt of 3-pιcolyl chloride (0.081 mmol) . After stirring 5 minutes triethylamine (300 uL, mmol) was added. The reaction was then stirred for 36 hours the reaction was quenched by the addition of 1.0 mL of saturated aqueous sodium bicarbonate. The reaction mixture was then diluted by the addition of 10 L of diethyl ether and washed by 2 x 10 mL of each saturated aqueous sodium bicarbonate and saturated aqueous brme. The combined organic portions were then dried over MgS04, filtered and concentrated in vacuo to provide the crude product. Purification of the crude solid was carried out by silica gel chromatography (1000 uM Sι02 prep, plate) elut g with 20 % Me0H/CH2Cl2. This provided 3.1 mg of the desired product 2, with 96 % purity by HPLC. The overall yield for addition, deprotection of N-Boc and coupling with 3-pιcolyl chloride was 9 I .
Example 27 Synthesis of Compound 3 A.
Figure imgf000185_0001
Allyl urea 1 (195.2 mg, 0.09 mmol) was dissolved 6.0 mL of DMF and cooled to 0 °C. This was followed oy the addition of NaH (54 mg, 1.0 mmol) . The glycidyl tosylate (410 mg, mmol) was then added as a solid. The reaction was stirred for 4 hours at 25 °C and then quenched by the addition of 4 L of saturated aqueous sodium bicarbonate. The reaction was then extracteα by 10 mL of Et20. The organic layer was then washeα oy 10 L of saturated aqueous sodium bicarbonate and 2 x 10 mL of saturated brme. The combined organic portions were then dried over MgS04, filtered and concentrated m vacuo to provide the desired epoxide 2 (180 mg, 73 % yield) . The epoxide was then used without further purification.
B .
Figure imgf000186_0001
Piperazine 1 (25.7 mg mmol) and epoxide 2 (22.6 mg, mmol) were heated to 75 °C in 1.5 mL of l-PrOH fcr 18 hours. After cooling to 25 °C the crude reaction mixture was concentrated in vacuo. Complete consumption of the epoxide was apparent by both tic and 1H NMR.
Figure imgf000186_0002
The Boc protected piperazine 1 from the previous step was stirred for 1.5 hours in 1.0 L of 4 N HCl in dioxane. This was followed by concentration in vacuo. The crude hydrochloride salt was then dissolved in 10 mL of CH2C12 and washed by 10 mL of both saturated sodium bicarbonate and saturated brine. The organic portion was then dried over MgS0 , filtered and concentrated in vacuo. The free amine was then taken up in 1 mL of DMF. This was followed by the addition of 3-picolyl chloride HCl salt (50 mg, mmol) and triethyl amine (300 uL) , respectively. The reaction was then stirred at 25 °C for 30 hours. The reaction was then quenched by the addition of 2 mL of saturated sodium bicarbonate and diluted by 10 L of Et20. The organic portion was then washed by 10 mL of saturated sodium bicarbonate and 2 X 10 L of saturated brine. The combined organic portions were then dried over MgS04, filtered and and concentrated in vacuo. The crude material was purified by silica gel chromatography (1000 uM prep, plate) eluting with 3:1, CH2Cl2:MeOH to provide 8.8 mg of the desired product 2. The overall yield for addition, deprotection of the N- Boc and reaction with 3-picolyl chloride was 19.3%.
Example 28 Synthesis of Compound 62
Figure imgf000187_0001
The THF lactam 1 (0.4 mmol) was dissolved in dry DMF at 0 °C and to this solution was added sodium hydride (0.47 mmol) . After 30 min of stirring, (s)- epichlorohydrin (0.47 mmol) was added and the reaction was allowed to warm to room temperature and stir overnight. The reaction was then diluted with water and extracted with ethyl acetate. The organic layer was washed sequentially with 0.5N HCl, saturated NaHC03 and brine, followed by drying (MgS0 ), filtration and concentration in vacuo to yield product (118 mg, crude) which was used as is. The lactam-epoxide (0.4 mmol, crude) was dissolved m isopropanol (2 mL) , and to this solution was added decahydroisoquinoline t-butylamide (0.7 mmol) . The mixture was then heated to 80 °C and stirred overnight. The reaction mixture was cooled and concentrated to dryness in vacuo, the residue of which was applied to a preperative TLC plate and eluted with 100% ethyl acetate to yield pure product (88 mg, 42%) as a mixture of diastereomers.
Example 29
Synthesis of Compound 92
A.
Figure imgf000188_0001
A stirred, cooled (-78 °C ) solution of 1.4 g (5.0 mmol) of pyrrolidinone in 35 mL of anhydrous tetrahydrofuran was treated in a dropwise fashion with 3.6 mL (7.2 mmoL) of lithium dusopropylamide. The resultant solution was stirred for 70 mm, and subsequently treated with 0.57 mL (6.0 mmoL) of 3- pyridine carboxaldehyde. The homogenous solution was allowed to ambiently warm to room temperature (RT) , and stirring was continued overnight. The reaction mixture was diluted with 400 mL of dichloromethane, washeα once with 150 mL of water, dried (magnesium sulfate), filtered, concentrated, and purified on silica gel using 3:1 ethyl acetate/hexanes as the eluent, affording 0.6 g (46%) of the desired compound as a golden oil which solidified upon standing.
λE NMR (d6-DMSO, 400MHz) 8.65 (s, IH) ; 8.47 (m, 2H) ; 7.83 (d, J = 8.0 Hz, IH) ; 7.41 (m, IH) ; 7.23 (m, 5H) ; 7.03 (t, J = 2.7 Hz, IH) ; 3.96 (m, IH) ; 3.07 (m, IH) ; 2.89 - 2.65 ( series of m, 3H ) . M+H (265.2) .
B ,
Figure imgf000189_0001
A vigorously stirred suspension of 330 mg ( 1.25 mmoL of eneamide and 80 mg of 10% palladium on carbon (Degussa) in 12mL of anhydrous methanol was hydrogenated (Hydrogen balloon) for 1 h. The mixture was diluted with 100 L of methanol, carefully filtered, concentrated, and purified on silica gel using ethyl acetate as the eluent, affording 295 mg (89%) of an isomenc mixture of the desired compounds as a golden oil which solidified upon standing.
1H NMR (d6-DMS0, 400MHz) 8.36 (s, 2H) ; 7.88 (s, IH) ; 7.56 (d, J = 7.9 Hz, IH) ; 7.27 - 7.12 (m, 7H) ; 3.66 (m, IH) ; 2.96 - 2.37 (series of m, 7H) . M+H (267.2); M+Na (289.2)
Figure imgf000190_0001
cis-isomer 2
The lactam obtained above was coupled to the corresponding epoxide according to the protocol used for Example 24. The final purification was performed on silica gel (2% 2M ammonia- ethanol in dichloromethane) to give the cis- and the trans- actam diastereomers each as white solids.
trans-isomer 1: Rf: 0.20 1H NMR (CDC13, . 400MHz) : 1.62(2H,m), 1.86(4H,m), 2.19(lH,m), 2.63(2H,m), 2.78- 3.10(8H,m), 3.65(lH,m), 3.75(lH,bt), 3.95(lH,t), 4.27(lH,t), 5.24(lH,m), 6.32(lH,d), 7-7.4 (14H,m) , 8.22(lH,s) , 8.34(lH,s) . M+H (604)
cis-isomer 2: Rf: 0.18. H NMR (CDC13, . 400MHz) 1.35(lH,m), 1.60(2H,m), 1.95(2H,m), 2.19(lH,dd), 2.48(lH,dd) , 2.60(lH,m), 2.8-3.05 (5H, m) , 3.10(lH,dd) , 3.26(lH,dd), 3.60(lH,m), 3.78(lH,m) , 3.99(lH,m) , 4.15(lH,bs), 4.24(lH,t) , 5.24(lH,m) , 6.18(lH,d) , 7.02(2H,d) , 7-7.3(10H,m) , 7.41(lH,d), 8.25(lH,s) , 8.40 (lH,d) . M+H (604)
Example 30
Figure imgf000191_0001
The iodolactam 1 (0.43 mmol) was dissolved in dry acetonitrile in a high pressure tube and to this solution was added diisopropylethylamme (Pierce, 0.65 mmol) followed by aniline 2 (Aldrich, 0.47 mmol) . The tube was sealed and the reaction heated to 70 °C with stirring overnight. The reaction was cooled to ambient temperature, solvent removed in vacuo, and the residue taken up in ethyl acetate/water. The organic layer was washed sequentially with saturated aqueous NaHC03 and brme, followed by drying (MgS04), filtration and concentration in vacuo. The crude residue was purified by flash silica gel chromatography eluting with 1:1 ethyl acetate/hexanes to give 61 mg of product 3; TLC Rf = 0.29 (1:1 ethyl acetate/hexanes); HPLC Rt = 12.6 mm (96%) ; MALDI-TOF MS m/z 267 (M+) . Example 31
A.
Figure imgf000192_0001
PMB lactam 1 (1.5 g, 5.07 mmol) was dissolved in THF (12 mL) , cooled to -78 °C, and to this solution /as added LDA (6.6 mmol , 1.3 eq.), over 7 minutes to give a greenish-brown anion. The reaction mixture was stirred at -78 °C for 55 minutes after which a solution of bromoacetonitrile (400 ul, 0.75 mmol, 1.1 eq.) was added over 2 minutes while keeping the internal reaction temperature at <-65 °C. The reaction was stirred at -78 °C for 2 hours, then warmed to room temperature and stirred for an additional 16 hours. The reaction was cooled to -50 °C and quenched with saturated ammonium chloride solution. The reaction was partitioned between ethyl acetate and a saturated bicarbonate solution. The aqueous layer was extracted with ethyl acetate. The combined organic layers were then washed with water, brme and dried (MgS04) and filtered. Concentration m vacuo afforded 1.6g of crude material, which was purified by silica gel chromatography to give 640 mg (38%) of the desired material 2.
1H NMR (CDCI3) d 7.31 (m, 3H) , 7.18 (d, 2 H) , 7.09 (d, 2H), 6.90 (d, 2H) , 5.08 (d, IH) , 3.92 (d, IH) , 3.81 (s, 3H) , 3.70 (m, IH) , 2.92 (dd, IH) , 2.72 (m,2H), 2.55 (dd,lH) , 2.42 (m, IH) , 2.19 (dd,lH) , 1.81 (m, IH) .
B,
Figure imgf000193_0001
PMB lactam 1 (640mg, 1.9 mmol) was dissolved in CH3CN (9 mL) . 1 L of water was added followed by 3.1 g of cerium ammonium nitrate. The reaction went from dark amber to light orange withm 5 minutes and was stirred at room temperature for 18 hours. The reaction was concentrated m vacuo and the residue was partitioned between ethyl acetate and a saturated bicarbonate solution. The aqueous layer was extracted with ethyl acetate. The combined organic layers were then washed with saturated bicarbonate solution, water, brme, dried (MgS04) and filtered. Concentration in vacuo afforded 590 mg of crude material, which was purified by silica gel chromatography (9:1 CH2C12 : EtOAc) to give 285 mg (70%) of the desired material 2. HPLC suggests 2 diastereomers, retention time 9.95 mm. (major) and 10.17 mm. (minor) . XH NMR (CDC13) d 7.37 (m, 2H) , 7.28 (m, 1 H) , 7.20 (m, 2H), 5.74 (br s, IH) , 3.95 (m, IH) , 2.85 (dd, IH) , 2.79-2.65 ( , 3H) , 2.55 (dd, IH) , 2.27 (m,2H). Example 32
A.
Figure imgf000194_0001
1
The PMB lactam 1 (0.46 mmol) was dissolved in dry THF at -78 °C and to this solution was added lithium diisopropylamide (Aldrich, 1.5 M in cyclohexane, 0.65 mmol). The solution was stirred for 15 minutes at -78 °C and 4- (Chloromethyl)-3, 5-dιmethylιsoxazole 2 (Acros Organics, 0.56 mmol) was added. The cooling bath was removed and the solution warmed to room temperature and stirred overnight. The reaction was diluted with water and extracted with ethyl acetate. The organic layer was washed sequentially with saturated aqueous NaHC03 and brine, followed by drying (MgS04), filtration and concentration in vacuo . The crude residue was purified by flash silica gel chromatography elutmg with 10% diethyl ether/dichloromethane to give 53 mg of product 3 as a mixture of diastereomers. B .
Figure imgf000195_0001
Lactam 1 (0.13 mmol) was dissolved in 7:3 acetonitrile/water. Ceric ammonium nitrate (Aldrich, 0.26 mmol) was added and the mixture was stirred at ambient temperature until the starting material was no longer evident by TLC. Acetonitrile was removed in vacuo, and the residue taken up in ethyl acetate/water. The organic layer was washed sequentially with saturated aqueous NaHC03 and brine, followed by drying (MgS04), filtration and concentration in vacuo . The crude residue was purified by flash silica gel chromatography eluting with 8% MeOH in dichloromethane to give 21 mg of product 2; TLC Rf = 0.47 (8% MeOH/CH2Cl2) .
Example 33
A .
Figure imgf000196_0001
Lactam 1 (1.43 mg, 4.86 mmol) was dissolved in anhydrous THF (25 mL) and cooled to -78 °C. This was followed by the addition of 3.9 mL of LDA (5.83 mmol,
1.2 eq.) . The anion solution was stirred at -78 °C for 45 minutes and then cannulated into a -78 °C solution of p-formaldehyde (437 mg) in 25 mL of THF, washing with 1 mL of THF. The reaction was warmed to room temperature over 4 hr and stirred overnight. The reaction was quenched by the addition of 10 L of a saturated sodium bicarbonate, and concentrated in vacuo to remove the THF. The crude reaction mixture was partitioned between ethyl acetate and saturated sodium bicarbonate. The aqueous layer was extracted with ethyl acetate. The combined organic layers was then washed with water, brine and purified by silica gel chromatography (gradient of 50 to 75 % ethyl acetate: hexanes), to provide 584 mg (45%) of the desired alcohol, as well as 265 mg of recovered starting material . The alcohol (316mg, 0.979 mmol) was then dissolved in 3 is of CH2C12 and added to a 0 °C solution of tnphenyl phosphine (734 mg, 2.8 EQ.) and NBS (534 mg, 3 EQ.) 3 mis of CH2C12• After 1 hour the reaction was quenched by the addition of 10 mL of Et20. The organic layer was then filtered and the filtrate washed with saturated sodium bicarbonate, brme, dried (MgS04) and filtered. Concentration m vacuo afforded the crude product which was purified by silica gel chromatography (CH2C12) to provide 151 mg (40% of the bromide.
The bromide (87.2 mg, 0.28 mmol) was dissolved in 2 mL of benzene and treated with imidazole (46mg, 3 EQ.). After heating to 125 °C for 20 hours the reaction was cooled to 25 °C and concentrated m vacuo . The crude product which was purified by silica gel chromatography (5 % MeOH/CH2Cl2) , to provide the addition product (50%) and the elimination product (2) in a 50 % yield.
B .
Figure imgf000197_0001
The lactam 1 (621 mg, 2.02 mmol) was dissolved in 7 L acetonitrile, followed by the addition of H20 (3 mL) . This was followed by the addition of CAN, 3.32 g (6.06 mmol, 3 EQ.) . The reaction was stirred at 25 °C for 1 hour. After concentrating the reaction in vacuo, the crude material was resuspended in ethyl acetate and washed with saturated sodium bicarbonate, brine, dried (MgS04) and filtered. Concentration in vacuo afforded the crude product which was purified by silica gel chromatography (3% methanol :CH2C12) to procide the desired unprotected lactam (122 mg, 32 %)
The , β-unsaturated lactam (55 mg, 0.29 mmol) was then heated to 130 °C in 2 ml of benzene containing imidazole (30 mg, 0.44 mmol) for 24 hours. After cooling to 25 °C, the reaction mixture was concentrate in vacuo . The crude material was purified by silica gel chromatography, eluting with 5% methanol :CH2C12 to provide 46.7 mg of the desired addition product (63 %) as well as 15.7 mg of recovered starting olefin (29 %) .
Example 34
Figure imgf000198_0001
The iodolactam 1 (0.45 mmol) was dissolved in dry acetonitrile in a high pressure tube and to this solution was added diisopropylethylamme (Pierce, 1.35 mmol) followed by indoline 2 (Aldrich, 0.54 mmol) . The tube was sealed and the reaction heated to 70 °C with stirring overnight. The reaction was cooled to ambient temperature, solvent removed m vacuo, and the residue taken up in ethyl acetate/water. The organic layer was washed sequentially with saturated aqueous NaHC03 and brme, followed by drying (MgS04), filtration and concentration m vacuo . The crude residue was purified by flash silica gel chromatography elutmg with ethyl acetate to give 113 mg of product 3; TLC Rf - 0.39 (ethyl acetate); HPLC Rt = 13.1 mm (92%) ; MALDI-TOF MS m/z 293 (M+) .
Example 35
A.
Figure imgf000199_0001
1
In an oven-dried 100 mL round-bottomed flask, the vmyl sulfone PMB lactam 1 (1.2126 g, 2.55 mmol) was dissolved in 50 mL of C6H6. Phenyl isocyanate (2.0 mL,
18.4 mmol) was added via syringe followed by the dropwise addition of nitroethane (0.4 mL, 5.56 mmol) .
Triethylamine (2.0 mL, 14.3 mmol) was added dropwise. The solution was refluxed for 15 minutes and cooled. A white solid precipitated durmg the heating period.
The mixture was cooled, poured into water and extracted with CH2C12_ The organic extract was dried (MgS0 ) anc3 evaporated in vacuo to afford a brown oil that was chromatographed to afford the isoxazole PMB lactam 2 (901 mg, 90%) as a light yellow oil.
B.
Figure imgf000200_0001
1 2
In a 25 mL round-bottomed flask, isoxazole PMB lactam 1 (900 g, 2.30 mmol) was dissolved in 14 mL of 70% CH3CN-H20. Ceric ammonium nitrate (3.607 g, 6.58 mmol) was added forming a dark orange solution. The mixture was stirred until the starting material was no longer evident by TLC (10% EtOAc/CH2Cl2) . The light yellow solution was diluted with CH2C12 and washed with water. The organic layer was separated, dried (MgS04), and evaporated in vacuo to afford a brownish-red oil that was chromatographed (10% EtOAc/CH2Cl2) to produce the lactam 2 (300.3 mg, 48%) as a colorless oil.
Example 36
Figure imgf000201_0001
The iodolactam 1 (0.78 mmol) was dissolved in dry acetonitrile m a high pressure tube and to this solution was added diisopropylethylamme (Pierce, 2.35 mmol) followed by N-methylanilme 2 (Aldrich, 0.94 mmol) . The tube was sealed and the reaction heated to 70 °C with stirring overnight. The reaction was cooled to ambient temperature, solvent removed m vacuo, and the residue taken up m ethyl acetate/water. The organic layer was washed sequentially with saturated aqueous NaHC03 and brme, followed by drying (MgS04), filtration and concentration m vacuo . The crude residue was purified by flash silica gel chromatography eluting with 2:1 ethyl acetate/hexanes to give 134 mg of product 3; TLC Rf = 0.24 (2:1 ethyl acetate/hexanes); HPLC Rt = 12.7 mm (80%) ; MALDI- TOF MS m/z 282 (M+) . Example V
A .
Figure imgf000202_0001
NaH (0.96 g, 40 mmol) was suspended into 20 L of dioxane. This was followed by the addition of diethyl malonate (4.6 mL, 40 mmol), then phenyl iodide (2.2 mL, 20 mmol) and finally copper (I) iodide (7.6g, 40 mmol). The reaction was then heated to 100 CC for 14 hours. The reaction was then quenched with water and diluted with ethyl acetate, the organic layer was washed with water and saturated NaCl, dried (MgS04) and concentrated in vacuo . The crude product was further purified by MPLC (Si02) eluting with 4:1, toluene: ethyl acetate to provide 1.21 g of product (29 % isolated yield) .
B .
Figure imgf000202_0002
CSCO3 MeCN
Figure imgf000202_0003
The alkylated malonic ester (1, 227 mg, 1.09 mmol) was stirred for 14 hours in acetonitrile (2.5 mL) containing; cesium carbonate (710 mg, 2.18 mmol) and the bromide (516 mg, 1.31 mmol) . The reaction was then concentrated to dryness in vacuo . After re-suspension of the reaction mixture in ethyl acetate, the reaction mixture was washed with water, saturated NaHC03 and saturated NaCl, dried (MgS04) and concentrated m vacuo . The crude product was further purified by MPLC (Sι02) to provide 200 mg of the desired product 35.2 % yield) .
H2 ( approx. 1 ATM)
Figure imgf000203_0001
Pd/C, HCl, EtOH EtOOC 0
Figure imgf000203_0002
To the malonate (1, 200 mg) in ethanol (3 mL) was added concentrated HCl (lOOuL) and an excess of 5% Pd / C (approx. 50 mg) . The reaction was then fitted with a balloon of H2 and hydrogenated for 14 hours. After purging the reaction mixture of H2 triethylamine (1 mL, 7 mmol, excess) and an excess of solid NaHC03 was added. After stirring for 30 minutes the reaction was filtered and concentrated m vacuo . The yellow oil was then re-dissolved m ethyl acetate and the reaction mixture was washed with water, saturated NaHC03 and saturated NaCl, dried (MgS04) and concentrated m vacuo to provide the desired product. The H NMR was consistent with the desired material.
Example 38
A.
Figure imgf000204_0001
In an oven-dried 25 L round-bottomed flask, the PMB- lactam 1 (563.7 mg, 2.75 mmol) was dissolved m 10 mL of THF. The solution was cooled to -78 °C and 1.5M LDA (2.0 mL, 3.00 mmol) was added dropwise via syringe producing the yellow color of the enolate. The solution was stirred for 15 minutes at -78 °C and propargyl bromide (310 uL, 3.48 mmol) was added dissipating the yellow color. The cooling bath was removed and the solution was warmed to room temperature and stirred overnight. The solution was poured into IN HCl and extracted with CH2C12. The organic extracts were combined and washed with saturated aqueous NaHC03. The organic layer was separated, dried (MgSθ4> and evaporated in vacuo to afford a brown oil that was cnromatographed (90% CH2Cl2/hexane) to produce the propargyl lactam 2 (577 mg, 86%) as a colorless oil. B.
Figure imgf000205_0001
In a 25 mL round-bottomed flask, propargyl PMB lactam 1 (358.2 g, 1.08 mmol) was dissolved in 6 mL of 70% CH3CN-H2O. Ceric ammonium nitrate (1.321 g, 2.41 mmol) was added forming a dark orange solution. The mixture was stirred until the starting material was no longer evident by TLC (10% EtOAc/CH2Cl2) . The light yellow solution was diluted with EtOAc and washed with water. The organic layer was separated, dried (MgS04) , and evaporated in vacuo to afford a yellow oil that was chromatographed (10% EtOAc/CH2Cl2) to produce the propargyl lactam 2 (145 mg, 63%) as a colorless oil.
Example 39
Figure imgf000205_0002
1
The iodolactam 1 (1.38 mmol) was dissolved in dry acetonitrile in a high pressure tube and to this solution was added diisopropylethylamine (Pierce, 4.15 mmol) followed by tetrahydroquinoline 2 (Aldrich, 1.66 mmol) . The tube was sealed and the reaction heated to 70 °C with stirring overnight. The reaction was cooled to ambient temperature, solvent removed in vacuo, and the residue taken up .in ethyl acetate/water. The organic layer was washed sequentially with saturated aqueous NaHC03 and brine, followed by drying (MgS04) , filtration and concentration in vacuo . The crude residue was purified by flash silica gel chromatography eluting with 1:1 ethyl acetate/hexanes to give 233 mg of product 3; TLC Rf = 0.21 (1:1 ethyl acetate/hexanes); HPLC Rt = 14.0 min (85%) ; MALDI- TOF MS m/z 307 (M+) .
Example 40
A.
Figure imgf000206_0001
1
In an oven-dried 250 mL round-bottomed flask, N- chlorosuccinimide (2.5177 g, 18.9 mmol) was dissolved in 75 mL of CH2C12. The solution was cooled to 0 CC and thiophenol (1.90 mL, 18.5 mmol) was added dropwise via syringe causing an immediate formation of a yellow color and an exotherm. The orange solution of PhSCI was stirred for 30 minutes at room temperature and a solution of the allyl lactam 1 (6.156 g, 18.4 mmoP was added dropwise dissipating the orange color. The light yellow solution was stirred for two hours and the solvent was removed in vacuo. CCI4 was added to the yellow oil that remained and the undissolved succinimide was removed by filtration. The filtrate was evaporated in vacuo to afford the diastereomeric chlorosulfides as a yellow oil that was chromatographed (CH2C12) rapidly to remove low Rf impurities. The two highest R spots were the chlorosulfide diasteromers .
The purified mixture of chlorosulfides was dissolved in CH2C12 and m-chloroperbenzoic acid (2.0 g, 11.6 mmol) was added with cooling from an ice-bath. The mixture was stirred for 10 minutes and filtered. The filtrate was evaporated in vacuo to afford a yellow oil (8.125 g, 86%) that produced two low Rf spots (CH2C1 ) using chin-layer chromatography for the two chlorosulfone diastereomers. The oil was redissolved in CH2C12 and DBU (2.7 mL, 18.1 mmol) was added dropwise at room temperature. The solution was heated for 15 minutes causing the solution to turn dark yellow. The solution was cooled and the solvent was evaporated in vacuo. The residue was chromatographed (CH2C12) to afford the pure vinyl sulfone 2 (4.805 g, 55%) as a colorless oil.
B .
Figure imgf000207_0001
In an oven-dried 25 mL round-bottomed flask, trimethylsilyl diazomethane (140 uL, 0.280 mmol) was dissolved in 5 mL of THF. The bright yellow solution was cooled to -78 CC and n-BuLi (320 uL, 480 mmol) was added. In a separate oven-dried 25 mL round-bottomed flask, the vinyl sulfone PMB lactam 1 (108 mg, 0.227 mmol) was dissolved in 5 mL of THF and added dropwise via syringe at -78 °C to the lithiate solution. The resulting solution was stirred for 1 hour at -78 °C and then two hours at 0- °C. The mixture was acidified with IN HCl and extracted with CH2C12. The organic extract was dried (MgS04) and evaporated in vacuo to afford a cloudy, colorless oil that was chromatographed (20% EtOAc/CH2Cl2) to produce the TMS pyrazole PMB lactam 2 (88.4 mg, 87%) as a clear, colorless oil.
C.
Figure imgf000208_0001
In an oven-dried 25 mL round-bottomed flask, the TMS pyrazole PMB lactam 1 (1.1345 g, 2.53 mmol) was dissolved in 110 mL of 91% CH3CN/H20. Tetrabutylammonium fluoride (2.7 mL of a 1.0M solution in THF, 2.70 mmol) was added dropwise via syringe. The reaction was refluxed for 48 hours and cooled. The solvent was evaporated in vacuo and the residue was dissolved in CH2C12. The organic solution was washed with IN HCl solution, dried (MgS04) , and evaporated m vacuo to afford a yellow oil that was chromatographeα (20% EtOAc/CH2Cl2) to afford the pyrazole (688 mg, 72%) as a light yellow oil.
D.
Figure imgf000209_0001
In an oven-dried 100 mL round-bottomed flask, the pyrazole PMB lactam 1 (588 mg, 1.57 mmol) was dissolved in 25 mL of THF. NaH (50 mg of a 60% dispersion in mineral oil, 2.08 mmol) was added. Gas evolution was observed. Methyl chloroformate (140 uL, 1.81 mmol) was added and the reaction was stirred at room temperature overnight. The mixture was acidified with IN HCl and extracted with CH2C1 . The organic extract was dried (MgS04), and evaporated in vacuo to afford the pyrazole carbamate PMB lactam 2 (588 mg, 87%) as a light yellow oil .
Figure imgf000209_0002
In an oven-dried 100 mL round-bottomed flask, the pyrazole carbamate PMB lactam 1 (577 mg, 1.33 mmol) was dissolved in 30 mL of 70% CH3CN-H20. Ceric ammonium nitrate (2.5123 g, 4.58 mmol) was added. The orange solution was stirred at room temperature until the starting material was no longer evident by TLC (1 hr) . The light yellow solution was poured into water and extracted with EtOAc. The organic extract was dried (MgS04) and evaporated vacuo to afford the pyrazole carbamate lactam 2 (228 mg, 55%) as a clear, colorless oil.
Example 41
Figure imgf000210_0001
In a heavy-walled screw-top test tube, the propargyl lactam 1 (1.111 g, 3.33 mmol) was dissolved in 7 mL of xylene. Tributyltm azide (1.965 g, 5.92 mmol) was added, the tube was sealed and heated to 205 °C overnight. The dark brown solution was cooled and directly chromatographed using a gradient from CH C12 to 50% EtOAc/CH2Cl2 to afford the triazole PMB lactam 2 (827 mg, 66%) as a light yellow oil. B.
Figure imgf000211_0001
In an oven-dried 100 mL round-bottomed flask, the triazole PMB lactam 1 (827 mg, 2.20 mmol) was dissolved in 40 mL of THF. NaH (124 mg of a 60% dispersion in mineral oil, 5.17 mmol) was added. Gas evolution was observed. Benzyl bromide (400 uL, 3.36 mmol) was added. The reaction was stirred at reflux until the starting material was not longer evident by thin-layer chromatography (50% EtOAc/CH2Cl2) . The mixture was acidified with IN HCl and extracted with CH2C12. The organic extract was dried (MgS04), evaporated m vacuo to afford a dark yellow residue that was chromatographed (20% EtOAc/CH2Cl2) to produce the benzyl triazole PMB lactam 2 (740 mg, 72%) as a light yellow oil.
C.
Figure imgf000211_0002
In an oven-dried 50 L round-bottomed flask, the benzyl triazole PMB lactam 1 (740 mg, 1.59 mmol) was dissolved 22 mL of 70% CH3CN-H20. Ceric ammonium nitrate (2.1 g, 3.83 mmol) was added. The orange solution was stirred at room temperature until the starting material was no longer evident by TLC (1 hr) . The mixture was poured into water and extracted with EtOAc. The organic extract was dried (MgS04) and evaporated vacuo to afford the benzyl triazole lactam 2 (336 mg, 61%) as a clear, colorless oil.
Example 42
Figure imgf000212_0001
BOC-lactam 1 (1.8 g, 6.6 mmol) was dissolved in THF (50 mL) and cooled to -78 °C. To this solution was added LDA (Aldrich, 1.5 M in cyclohexane, 5.3 mL, 7.9 mmol) via syringe over 10 minutes. After stirring for 60 mm at -78 CC, acetone (4.9 mL, 66 mmol) was added via syringe over 1 minute. The reaction was stirred for an additional 40 minutes before being quenched with IN HCl (15 mL) . Ethyl acetate (100 mL) was added and the layers were partitioned. The organic layer was washed with brine, dried over magnesium sulfate, filtered, and concentrated vacuo to a yellow oil that slowly crystallized. The crude alcohol was dissolved dichloromethane (50 mL) and Martin's sulfurane (Aldrich, 7.5 g, 11 mmol) was added in one portion. The reaction was stirred for 36 h at room temperature before being concentrated in vacuo. Flash chromatography over silica gel (3:1 hexane:ethyl acetate) provided the alkene as a mixture of isomers. The alkene, 10% Pd-C (1.0 g) , and methanol (40 mL) were combined in a Parr bottle and pressurized to 50 psi of hydrogen gas. After 4 h of agitation, the reaction vessel was evacuated and filtered through a plug of Celite. The cake was washed with ethyl acetate (20 mL) and the combined filtrate was concentrated in vacuo to give the isopropyl BOC-lactam as a pale yellow oil. The lactam was dissolved in dichloromethane (20 mL) and trifluoroacetic acid (10 mL) was added slowly. The reaction was stirred at room temperature for 24 h before being diluted with ethyl acetate (100 mL) and carefully neutralized with 10% sodium carbonate to pH 7. The layers were partitioned and the organic layer was dried over magnesium sulfate, filtered, and concentrated m vacuo. Flash chromatography over silica gel (3:1 ethyl acetate:hexane) gave the isopropyl lactam as a white powder. MS (ES+) = 240 (M+Na)
Example 43
Figure imgf000214_0001
-78°C~- RT
A stirred, cooled (-78 °C) solution of 1.4 g (5.0 mmol) of pyrrolidinone in 35 mL of anhydrous tetrahydrofuran was treated in a dropwise fashion with 3.6 mL (7.2 mmoL) of lithium diisopropylamide. The resultant solution was stirred for 70 mm, and subsequently treated with 0.57 mL (6.0 mmoL) of 3-pyrιdme carboxaldehyde. The homogenous solution was allowed to ambiently warm to RT, and stirring was continued overnight. The reaction mixture was diluted with 400 L of dichloromethane, washed IX with 150 mL of water, dried (magnesium sulfate) , filtered, concentrated, and purified on silica gel using 3:1 ethyl acetate/hexanes as the eluent affording 0.6 g (46%) of the desired compound as a golden oil which solidified upon standing. 1H NMR (d6-DMSO, 400MHz) 8.65 (s, IH) ; 8.47 (m, 2H) ; 7.83 (d, J = 8.0 Hz, IH) ; 7.41 (m, IH) ; 7.23 ( , 5H) ; 7.03 (t, J = 2.7 Hz, IH) ; 3.96 ( , IH) ; 3.07 (m, IH) ; 2.89 - 2.65 (series of m, 3H) . M+H (265.2) . Example 44
Figure imgf000215_0001
l
The first step of the sequence was performed as for Example 43. The olefin was carried forward as follows:
step 2 A vigorously stirred suspension of 330 mg (1.25 mmoL) of eneamide and 80 mg of 10% palladium on carbon (Degussa) in 12mL of anhydrous methanol was hydrogenated (Hydrogen balloon) for 1 h. The mixture was diluted with 100 mL of methanol, carefully filtered, concentrated, and purified on silica gel using ethyl acetate as the eluent affording 295 mg (89%) of an isomeric mixture of the desired compounds as a golden oil which solidified upon standing. IH NMR (d6-DMS0, 400MHz) d 8.36 (s, 2H) ; 7.88 (s, IH) ; 7.56 (d, J = 7.9 Hz, IH) ; 7.27 - 7.12 ( , 7H) ; 3.66 (m,
IH) ; 2.96 - 2.37 (series of m, 7H) . M+H (267.2) ; M+Na (289.2)
Example 45
Figure imgf000216_0001
The synthesis of the 2-pyridyl methylpyrrolidone was carried out as shown in -Example 44.
Example 46
Figure imgf000216_0002
The 4-pyridylmethylpyrrolidone was prepared following procedures outline for Example 44.
F.x am l e 47
Boc-Phenylalanmal
Figure imgf000217_0001
4 TFA, neutralize X=0, N-Bn
X = N-Bn
A .
A solution of 5.06g (20 mmol, 1 equiv) of tert-Butyl- P, P-dimethylphosphonoacetate m 15 mL THF cooled to 0 °C was treated with 0.528g of NaH at 0 °C and tnen warmed up to room temperature for 30 mm. Next, solution of 5.0g (20 mM, 1 equiv) of Boc-Phenylalaninal in 5 mL THF was added dropwise at 0°C and the reaction continued for 2 h. The crude product was diluted with ethyl acetate and partitioned with aqueous citric acid (2x) , sodium bicarbonate (2x) , organics collected and dried over magnesium sulfate. The product was then dissolved m 100 mL methanol, added 0.6g 10% Pd/C and hydrogenated at 25 psi overnight, and the desired compound purified on a silica column using 1/4 ethyl acetate/hexane. Yield 3.8g (51.4%) . 1H NMR (CDCL3, 300 MHz) δ (broad signals and conformational averaging) 7.20 (m, 5H) , 4.46 (m. 0.5H), 3.79 (m, 0.5H), 3.72 (s, 0.5H), 2.80 (m, 0.5H), 2.46 (m. 0.5H), 2.27 (m, IH) , 1.78 (m, IH) , 1.50 ( . IH) , (1.44, 1.42, 1.41, 1.38} (all s, total 18H) . Low resolution MS m/e 372.2 (M+Na+) . B .
A solution of 4.63 g (13.25 mmol, 1 equiv) of the above ester in 200 mL of THF was treated with 40 mL (39.75 mmol, 3 equiv) of IM lithium bis (trimethylsilyl) amide THF at -78 °C. After 90 mm at -78 °C, the solution was added 5.5g (13.25 mmol, 1 equiv) of N-benzyl-N- bis dodoethane) in 10 mL of THF and the reaction continued for 6 hours durmg which it reached the room temperature. The reaction was quenched with 10% aqueous solution of citric acid and extracted to ethyl acetate, and the product treated with 1:1 (v/v) DCM/TFA (40 mL) for 40 mm, after which solvents were removed and the crude purified to homogeneity by RP HPLC with total yield of 14.2%. The resulting TFA salt was then neutralized with triethylamine, extracted between ethyl acetate/water, organics collected and dried, thus yielding a free base form of the spiropyrrolidone product which is used in subsequent coupling to the epoxide. 1H NMR (TFA salt, CDCL3, 300 MHz) δ 7.30 (m, 10H), 5.85 ( , IH) , 4.16 ( , 2H) , 3.86 (m, IH) , 3.68 (m, IH), 3.36 (m, 3H) , 2.88 (dd, IH) , 2.62 (dd, IH) , 1.7-2.2 (m, 6H) . Low resolution MS m/e 335.2 (M+H+)
Example 48
Spirocycle X=0 was synthesized accordmg to bisalkylation protocol of Example 47 above except that bis-O- dodoethyl) ether was used in reaction step B (1.26g, 3,87 mmol, 1 equiv) . 1H NMR (d6-DMS0, 300 MHz) δ 7.79 (s, IH) , 7.22 (m, 5H) , 3.73 (m, 3H) , 3.24 (m, 2H) , 2.88 (dd, IH, J=4.8, 13.4), 2.57 (dd, IH, J=8.4, 13.4), 2.03 (m, IH) , 1.76 (m, IH) , 1.55 (m, 2H) , 1.22 (m, IH) , 1.01 (m, IH) . Low resolution MS m/e 246.2 (M+H+) .
Example 49
Boc-Phenylalaninal
Figure imgf000219_0001
=α$
Spiro cycle X=CH2
A.
A solution of 1.36g (3.88 mmol, 1 equiv) of the ester from Example 47 step A m 5 mL of THF was cooled to - 78 °C and treated with 9.32 mL (9.32 mmol, 2.4 equiv) of IM lithium bis (trimethylsilyl) amide m THF. After 1 h at -78 °C, 0.992g (4.27 mmol, 1.1 equiv) of 1,5- lodochloropentane was added and the reaction allowed to progress at -15 °C for 1 h, quenched with 10o aqueous citric acid, and extracted to ethyl acetate, resulting m 1.60g of product. Low resolution MS m/e 476.2 (M+Na+)
B.
A solution of 1.6g (3.53 mmol, 1 equiv) of the above chloride m 30 mL acetone was treated with 5.29 g (35.3 mmol, 10 equiv) of Nal and refluxed overnight. Solvents were then removed and the residue partitioned between ethyl acetate/water. Organics were dried with magnesium sulfate and purified on silica gel using 1/3 ethyl acetate/hexane resulting in 1.2 g of the desired iodide (62.4% yield after chromatography) .
•"H NMR (CDCL3, 300 MHz) δ 7.20 (m, 5H) , 4.38 ( , IH) , 3.79 (m, IH) , 3.13 (t, 2H, J=6.9), 2.73 (m, 2H) , 2.25
(m, IH), 1.76 (m, 2H), 1.43 (s, 9H) , 1.38 Ts, 9H) , 1.2-
1.7 (m, 7H) . Low resolution MS m/e 568 (M+Na+), m/e 362.2 (M+H+)
C. A solution 1.15g (2.1 mM, 1 equiv) of the above product in 20 mL of anhydrous THF was cooled to -78 °C and treated with 3.2 mL (3 mmol, 1.5 equiv) of IM lithium bis (trimethylsilyl) amide in THF. The reaction was then allowed to warm up to room temperature, solvents removed and the crude product purified on preparative HPLC. 1H NMR (CDCL3, 300 MHz) δ 7.32 (m. 4H) , 7.19 (d, 12H), 3.88 (m, IH) , 2.82 (m, 2H) , 2.24 (dd, IH) , 1.2-
1.8 (m, 11H) . Low resolution MS m/e 384.2 (M+Na+), m/e 362.2 (M+H+) .
Example 50
Figure imgf000220_0001
3. H2 , Pd-C A.
The Boc-pyrrolidone (4.4 g, 16 mmol) was dissolved in THF (40 mL) and cooled to -78 °C. To this solution was added LDA (Aldrich, 1.5 M in cyclohexane, 12.8 mL, 19 mmol) via syringe over 10 minutes. After stirring for 60 min at -78 °C, 3-formyl-5, 6-dihydro-2H-pyran (US Patent 4,532,337) (1.8 g, 16 mmol) in THF (5 mL) was added via syringe over 1 minute. The reaction was then allowed to reach room temperature and stir for 20 h before being quenched with saturated ammonium chloride (15 L) . Ethyl acetate (50 mL) was added and the layers were partitioned. The organic layer was washed with brine, dried over magnesium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash chromatography over silica gel (95:5 chloroform:methanol) to give dihydropyran lactam as a beige powder. MS (ES+) = 270 (M+l), 292 (M+Na)
B.
The dihydropyran obtained above (1.2 g, 4.4 mmol), 10% Pd-C (0.2 g) , and methanol (35 mL) were combined m a Parr bottle and pressurized to 50 psi of hydrogen gas. After 3 h of agitation, the reaction vessel was evacuated and filtered through a plug of Celite. The cake was washed with methanol (20 mL) and the combined filtrate was concentrated in vacuo. Flash chromatography over silica gel (95:5 chloroform:methanol) gave the tetrahydropyran lactam 2 as a white powder. MS (ES+) = 274 (M+l), 296 (M+Na) Example 51
Figure imgf000222_0001
A.
A solution of 2.6g (8.24 mmol, 1 equiv) of the allyl- pyrrolidone in 80 mL tetrahydrofuran and 25 mL water was cooled to 0 °C and treated with 5.29 g (24.7 mmol, 3 equiv) NaI04, followed by the addition of 838 mg of 2.5% solution of osmium tetroxide in 2-methyl-2- propanol. The reaction was continued for 2 h at room temperature, solvents removed and the residue partitioned between ethyl acetate and water. Ethyl acetate was then dried over MgS04, resulting in 3.0 g of the crude aldehyde.
1H NMR (CDCL3, 300 MHz) δ 9.75 (s, IH) , 7.22 ( , 5H) , 4.32 (m, IH), 3.05 (m, 2H) , 2.82 (m, 3H) , 2.53 (m, IH) , 2.22 (m, IH) , 1.58 (s, 9H) . Low resolution MS m/e 356.1 (M+Na+); m/e 689.3 (2M+Na+).
B.
A solution of 2.88g of the above aldehyde in 10 mL methanol was cooled to 0 °C and sodium borohydride was added over 2 h, until all the starting material (Rf=0.55, Merck Kiselgel 60, 0.25 mm, 1:1 ethyl acetate/hexane) was consumed. The title compound had Rf=0.30 (same conditions). Solvents were then removed, and the residue was extracted between ethyl acetate and 10% aqueous citric acid. Organic fractions were washed with water and dried over magnesium sulfate. Purification on a silica column (1:1 ethyl acetate/hexane) afforded 1.5 g (57% yield) of the alcohol. Low resolution MS m/e 342.2 (M+Na ) ; m/e 661 . A (2M+Na+) .
C.
A solution of 0.46g (1.44 mmol, 1 equiv) of the above alcohol in 4 mL tetrahydrofuran was treated with 0.215 g (1.875 mmol, 1.3 equiv) of mesyl chloride and 0.242g (1.875 mmol, 1.3 equiv) of diisopropylethylamme. The reaction was allowed to proceed for 30 mm at room temperature, solvents removed and the residue partitioned between ethyl acetate and water. Organics were dried with magnesium sulfate and purified on a silica column (1/1 ethyl acetate/hexane), yielding 0.50 g (87.3%) of the desired mesylate. Rf=0.57 (Merck Kiselgel 60, 0.25 mm, 1:1 ethyl acetate/hexane) . A NMR (CDCL3, 300 MHz) δ 7.22 (m, 5H) , 4.39 (m, 3H) , 3.09 (dd, IH, J=6.4, 13.2), 2.98 (s, 3H) , 2.76 (dd, IH, J=8.9, 13.2), 1.64 (m, 2H) , 1.57 (s, 9H) .
D.
A solution of 0.33g (0.831 mmol, 1 equiv) of the above mesylate in 3 mL DMF was cooled to 0 °C and treated with 26 mg (1.080 mmol, 1.3 equiv) of sodium hydride. After 3h at room temperature the reaction was quenched with aqueous citric acid and purified on silica gel using 1:3 ethyl acetate/hexane (v:v) . The resulting product (0.18g, 72.0% yield) was then treated with 1:1 dichloromethane/ trifluoroacetic acid (5 ml) for l/2h, re3u_tmg in 0.12g (71.8%, based on mesylate) of the dt 3d product. 1H NMR (CDCL3, 300 MHz) δ 7.23 (m, 5H) , 7.04 (broad s, IH) , 3.99 (m, IH) , 2.85 ( , 2H) , 2.26 (dd, IH, J=8.1, 12.9), 1.92 (dd, IH, J=5.0, 12.9), 1.10 ( , 2H) , 0.72 (m, 2H) . Low resolution MS m/e 342.2 (M+Na+);
Example 52
Figure imgf000224_0001
A solution of 1.5g (5.4 mMol) of the pyrrolidinone in 25 mL of tetrahydrofuran was cooled to -78 °C and treated with 4.3 mL (6.5 mMol) of lithiumdnsopropyl amide (2M THF). After stirring for 0.25h, acetone (2.8g (50 mMol) was added, the reaction mixture was kept at -78 °C for 2 h and then quenched with IN hydrochloric acid. Extraction with etnyl acetate, drying over magnesium sulfate and removal of the solvent in vacuo afforded the crude product which was redissolved in 25 mL of dichloromethane and treated with 8g of Martin's sulfurane. After stirring for 12h at -5 °C, the mixture was participated between ethyl acetate and IN hydrochloric acid. Drying over magnesium sulfate and removal of the solvent gave the desired alkene. 0.755 g of the crude alkene were dissolved in 15 L of toluene and treated with 3 mL (3 mMol) of iethyl alum umcyanide (lm in toluene) and the resulting mixture was stirred at 25 °C for 5 h. The solvent was removed and the residue was chromatographed on silica gel (20% ethylacetate- hexanes) to give the desired nitrile (0.4g) as a colorless oil. Deprotection with trifluoroacetic acid- dichloromethane (1:1) for 3h at 25 °C followed by chromatography on silica gel gave the desired lactam (0.22g) as a white solid. M+H: 243
Example 53
Figure imgf000225_0001
A. A solution of 3-ιodo-5-benzyl-pyrrolιdmone (2.67 g, 8.87 mmol) and sodium azide (0.69 g, 10.61 mmol) dimethylformamide (20 L) was stirred at ambient temperature under a nitrogen atmosphere for 18h. The solvent was evaporated using a stream of nitrogen, and the residue was dissolved in ethyl acetate, washed with water and brine, and concentrated m vacuo to give a yellow solid. Chromatography on silica gel, elutmg with hexane:ethyl acetate (4:1), gave 1.82 g of the product as a 1:1 mixture of diastereomers which was used without separation in the next reaction. MS:
ES+, 239 (M+Na). The chromatography also gave 0.12 g of the trans isomer as a colorless oil and 0.43 g of the cis isomer as a colorless oil which crystallized upon standing. TLC (hexane : ethyl acetate (1:1) ) Rf trans isomer =0.6 and Rf cis isomer = 0.5.
B.
A mixture of the above azide (0.575 g, 2.66 mmol) and 5% palladium on carbon (0.030 g) in methanol (20 L) was stirred under 40 psi of hydrogen for 18h at ambient temperature. The mixture was filtered through a pad of Celite to remove the catalyst, followed by filtration through 5 g of silica gel, washing with chloroform:methanol (9:1) . The filtrate was concentrated m vacuo to give 0.46 g (90%) of the product as a mixture of diastereomers. MS: ES+, 191 (M+l) and 213 (M+Na) .
C. A solution of the above amme (0.44 g, 2.3 mmol), 4- anisylchlorodiphenylmethane (0.71 g, 2.3 mmol) and triethylamine (0.5 L, 3.5 mmol) m dichloromethane (20 mL) was stirred under a nitrogen atmosphere at ambient temperature for 18h. The solution was washed with water (2x50 mL) and brine, dried (MgSθ4), anc* concentrated m vacuo . The residue was purified by chromatography on silica gel, eluting with hexane:ethyl acetate (7:3) then with hexane: ethyl acetate (1:1), to give 0.41 g of the cis isomer as a yellow solid and 0.19 g of the trans isomer as a white solid. TLC
(hexane : ethyl acetate (7:3)) Rf cis isomer =0.5 and Rf trans isomer = 0.4. Example 54
Figure imgf000227_0001
Iodolactam 1 (prepared as described previously in Example 7) (0.55 g, 1.8 mmol) was dissolved in DMF (5 mL) and treated with 2-fluoroanilme (Aldrich, 0.20 g, 1.8 mmol) and solid sodium carbonate (0.39 g, 3.7 mmol) . The reaction was then heated to 70 °C for 24 h before the solvent was removed in vacuo. Ethyl acetate (50 mL) and water (20 mL) were added and the layers were partitioned. The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo.
Flash chromatography over silica gel (1:1 hexane:ethyl acetate) gave the anilinolactam 2 as a pale yellow foam. MS (AP+) = 285 (M+l), 307 (M+Na)
Exampl e 55
Figure imgf000227_0002
Using the procedure described in Example 54, the anilinolactam was prepared, purified, and isolated as a beige foam. MS (AP+) = 285 (M+l), 307 (M+Na)
Example 56
Figure imgf000228_0001
Using the procedure described in Example 54, the anilinolactam was prepared, purified, and isolated as a beige foam. MS (AP+) = 292 (M+l), 314 (M+Na)
Example 57
Figure imgf000228_0002
Iodolactam 1 (prepared as described previously in Example 7) (0.77 g, 2.6 mmol) was dissolved in absolute ethanol (10 mL) and treated with 3-aminopyridine (0.26 g, 2.8 mmol) and solid sodium carbonate (0.40 g, 3.8 mmol) . The reaction was then heated at reflux for 24 h before the solvent was removed in vacuo. Chloroform (50 mL) and water (20 mL) were added and the layers were partitioned. The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo. Preparatory silica gel TLC (95:5 chloroform:methanol) gave the pyridylammolactam 2 as a red oil. MS (AP+) 268 (M+l), 290 (M+Na)
Example 58
A. KCN, DMF
Figure imgf000229_0001
1. 2.
A .
To a solution of iodolactam 1 (13.43 g, 44.6 mmol, 1 eq) in dimethylformamide (60 mL) under nitrogen was added potassium cyanide (3.49 g, 1.2 eq) . After stirring at ambient temperature for 24 h, the reaction mixture was evaporated in vacuo and the residue was partitioned between ethyl acetate, saturated aqueous brme and water. The layers were separated and the aqueous layer was back-extracted twice with ethyl acetate. The combined organic layers were washed with saturated aqueous brine, dried over anhydrous magnesium sulfate, filtered and evaporated in vacuo . The residue was purified by flash silica gel chromatography eluting with hexane : acetone (3:1) . Fractions containing the product were combined, evaporated in vacuo to provide 5.89 g (66%) of cyanolactam as a mixture of diastereomers. MS (APCI) : M+Na = 223.
B.
A solution of cyanolactam (5.78 g, 28.9 mmol) from step A absolute ethanol (233 mL) under Nitrogen was combined with 10 wt.% Palladium on charcoal (2.33 g) and concentrated hydrochloric acid (9.31 mL, 4 eq.) . The mixture was reduced under hydrogen gas at 50 psi for 16 h. The reaction was purged with nitrogen, filtered and evaporated in vacuo . The residue was combined with toluene (~ 100 mL) and concentrated m vacuo to a residue to remove residual water. The azeotropic removal with toluene was repeated four times leaving a residue which was dried under high vacuum to provide the crude amine as a gum (7.18 g, 103%) . MS (ESI) : M+l = 205.
C.
The crude amme (7.16 g, 29.8 mmol, 1 eq) from step B was combined under argon in dichloromethane (100 mL) with diisopropylethylamme (13 mL, 74.4 mmol, 2.5 eq) and triphenylmethylchoride (9.13 g, 32.7 mmol, 1.1 eq) . After stirring at ambient temperature for 16 h, the reaction mixture was treated with 5% w/v aqueous potassium carbonate and transferred to a separatory funnel. After separating the layers, the aqueous layer was back-extracted with dichloromethane and the combined organic layers were dried over anhydrous sodium sulfate and evaporated m vacuo to proved a crude mixture of diastereomers. The mixture was purified by flash silica gel chromatography eluting with ethyl acetate : hexane (3:7). Fractions containing the less polar diastereomer were combined and evaporated in vacuo to provide 3.52 g (26 %) of trityl protected amine as a crystalline solid. MS (APCI) : M+Na = 469.
Example 59
An alternate procedure for the synthesis of the benzyllactam:
A.
Figure imgf000231_0001
A mixture of methyl 2- (triphenylphosphoranylidene) - hydrocinnamate (13.20 g, 31.1 mmol, 1.15 eq) and N- tertbutoxycarobonyl-L-phenylalanal (6.76 g, 27.1 mmol, 1 eq) were combined in 200 mL chloroform and allowed to stir at ambient temperature over 64 h. The reaction was concentrated in vacuo and the residue was purified by flash silica gel chromatography eluting with 85:15 hexane : ethyl acetate. Fractions containing the product were combined and evaporated in vacuo to provide the olefin as a crystalline solid (9.38 g, 77%) . MS (ESI) : M + Na = 418. B .
Figure imgf000232_0001
A solution of the olefin (9.30 g, 23.5 mmol, leq) from step A in absolute ethanol (250 mL) was combined under nitrogen with Palladium on carbon (10 wt%, 1.90 g) and reduced under a balloon of Hydrogen gas over 16 h. The reaction mixture was purged with nitrogen, diluted with dichloromethane, filtered, and evaporated in vacuo to low volume. The solution was diluted with dichloromethane and filtered through a pad of diatomaceous earth washing with dichloromethane. The filtrate was evaporated in vacuo and dried under vacuum to provide a 5:1 mixture of diastereomers of the BOC- amino ester as an oil (9.68 g, 104 %) . MS (ESI) : M + Na = 420.
The oil was dissolved in dichloromethane (25 mL) and treated with trifluoroacetic acid (25 mL) under Argon. After stirring for 0.5 h at ambient temperature, the reaction mixture was evaporated in vacuo . The residue was dissolved in methanol (50 mL) and treated with diisopropylethyl amine (17 mL) followed by anhydrous potassium carbonate (13.49 g, 98 mmol, 4 eq) and stirred for 16 h at ambient temperature under an Argon atmosphere. The mixture was evaporated in vacuo and the residue was partitioned between dichloromethane and water. The layers were separated and the aqueous layer was back-extracted three times with dichloromethane. The combined organic layers were washed with aqueous hydrochloric acid (IN) and the layers were separated. The aqueous layer was back-extracted with dichloromethane and the combined organic layers were dried over anhydrous magnesium sulfate and evaporated in vacuo to a residue. The crude product was purified by flash silica gel chromatography eluting with a gradient of 45-60 % ethyl acetate in hexane. Fractions containing the less polar diastereomer were combined and concentrated in vacuo to a solid and dried under high vacuum to provide the enantiopure lactam as a white crystalline solid (4.48 g, 72%). MS (ESI): M + Na = 288. H NMR (CDC13) : 1.90 ( , IH) ; 2.01 (m, IH) ; 2.67 (m, 4H) ; 3.16 (m, IH) ; 3.65 (m, IH) ; 5.70 (s, IH) ; 7.18 (m, 10H) .
Example 60
Synthesis of Compound 123
Figure imgf000233_0001
Figure imgf000233_0002
To a suspension of (2S)- (+) -glycidyl 3- nitrobenzenesulfonate 1 (Aldrich, 19.47 mmol) and potassium carbonate (Baker, 38.93 mmol) in dry acetonitrile was added (S) -t-butyl decahydro-3- isoquinoline carboxamide 2 (NSC Technologies, 21.41 mmol) and the reaction stirred at ambient temperature overnight. The solvent was removed in vacuo, and the residue taken up in ethyl acetate/water, the organic layer was washed sequentially with saturated aqueous NaHC03 and brine, followed by drying (MgS04), filtration and concentration in vacuo . The crude residue was purified by flash silica gel chromatography eluting with 10% diethyl' ether/dichloromethane to give 3.62 g of product 3; HPLC Rt = 9.2 min (100%), TLC Rf = 0.26 (10% diethyl ether/dichloromethane); 1H NMR
(CDC13) d 6.59 (br s, 1 H) , 3.00 (d, 1 H) , 2.97 (m, 1 H) , 2.89 (dd, IH) , 2.73 (m, IH) , 2.65 (m, 1 H) , 2.57
(m, 1 H) , 2.22 (dd, IH) , 2.08 (dd, IH) , 1.81-1.70 ( , 4 H) , 1.65-1.19 (m, 8 H) , 1.38 (s, 9 H) .
B.
Figure imgf000234_0001
Phosphazene Base Rj-t-Bu H THF,-78°Ctor.t.
2-pyridylmethyl lactam 1 (35 mg, 0.13 mmol) was dissolved in anhydrous THF (1 L) and cooled to - 78 °C. Phosphazene Base P4-t-Bu (Fluka, 1.0M in hexane, 130 uL, 0.13 mmol.) was added to give an orangish brown anion. The anion solution was stirred at -78 °C for 35 minutes and was then cannulated under nitrogen over 30 seconds into a -78 °C solution of 2 (39 mg, 0.13 mmol) in ImL of THF and was washed in with 0.5 mL of THF. The reaction was gradually warmed to room temperature over 4 hr, then stirred at room temperature for 3 days. The reaction was cooled to - 78 °C, quenched with 0.5 mL of a saturated ammonium chloride solution, and concentrated in vacuo to remove the THF. The residue was partitioned between ethyl acetate and saturated bicarbonate solution and the aqueous layer was extracted with ethyl acetate. The combined organic layers were then washed with water, brine and dried (MgS04) and filtered. Concentration in vacuo afforded 75 mg crude material which was purified via silica gel to give 18 mg(25%) of 3. Maldi MS: M + H = 561.5 (MW = 560.79) . TLC (EtOAc) Rf = 0.19 (major diast.) & 0.29 (minor diast.) . TLC (5% MeOH/EtOAc) Rf = 0.28 (major diast.) & 0.36 (minor diast.) . HPLC retention times were 11.24 min. (major) & 11.32 mm. (minor) .
1H NMR (CDC13) d 8.52 (m, IH) , 7.61 (m, 1 H) , 7.34-7.10 (m, 7H) , 6.10-5.95 (m, IH) , 4.11 (m, IH) , 3.96-3.73 (m, 3H) , 3.46-2.74 (m, 6H) , 2.65-2.47 (m, 2H) , 2.23 (m, 2H), 2.10-1.15(m, 15H) , 1.37 (s,9H) .
Example 61
Synthesis of Compound 72
Figure imgf000236_0001
3-pyridylmethyl lactam 1 (85 mg, 0.32 mmol) was dissolved in DMF (1.5 mL) , cooled to 0 °C, and to this solution was added sodium hydride (0.48mmol) to give a yellow anion. The reaction mixture was stirred at 0 °C for 70 minutes after which (s) -epichlorohydrin (35 ul, 0.45 mmol) was added neat. The reaction was stirred at 0 °C for 5 minutes, then warmed to room temperature and stirred for 24 hours. The reaction was cooled to 0 °C and quenched with 0.5 mL of a saturated ammonium chloride solution. The reaction was partitioned between ethyl acetate and a saturated bicarbonate solution. The aqueous layer was extracted with ethyl acetate. The combined organic layers were then washed with water, brine and dried (MgS04) and filtered. Concentration in vacuo afforded 49 mg of crude epoxide which was used without further purification. B.
hPrOH
Figure imgf000237_0002
Figure imgf000237_0001
Crude lactam epoxide 1 (49 mg) and decahydroisoquinoline 2 (91 mg, 0.38 mmol) were heated to 65-70 °C in isopropanol. After 90 hours the reaction was cooled to 25 °C and stirred for 1 hour at room temperature. The reaction was then concentrated in vacuo, and purified by silica gel chromatography, eluting with 5 % MeOH : EtOAc, providing 30 mg (87% pure by HPLC) of desired product 3 as a mixture of 4 diastereomers. HPLC shows 2 split peaks 11.30 min. & 11.04 min.. TLC (5% MeOH/CH2Cl2) Rf = 0.27. TLC (10% MeOH/CH2Cl2) Rf = 0.45.
*H MR (CDC13) d 8.45-8.35 (m, 2H) , 7.48 (m, 1 H) , 7.35-7.09 ( , 6H) , 6.63-5.94 (m, IH) , 3.98-3.63 (m, 3H) , 3.42-2.73 (m, 5H) , 2.70-2.11 (m, 5H) , 2.07-1.20(m, 16H) , 1.36 (s, 9H) .
Example 62
Synthesis of Compound 54
Figure imgf000238_0001
4-pyridylmethyl lactam 1 (33 mg, 0.12 mmol) was dissolved in anhydrous THF (1 mL) and cooled to -78 °C. Phosphazene Base P4-t-Bu (Fluka, 1. OM in hexane, 125 uL, 0.125 mmol) was added to give a brown anion. The anion solution was stirred at -78 °C for 35 minutes and was then cannulated under nitrogen over 30 seconds into a -78 °C solution of 2 (39 mg, 0.13 mmol) in ImL of THF and was washed in with 0.5 mL of THF.
The reaction was gradually warmed to room temperature over 4 hr, then stirred at room temperature for 3 days. The reaction was cooled to -78 °C, quenched with 0.5 L of a saturated ammonium chloride solution, and concentrated in vacuo to remove the THF. The residue was then partitioned between ethyl acetate and saturated bicarbonate solution. The aqueous layer was extracted with ethyl acetate and the combined organic layers were then washed with water, brine and dried (MgS04) and filtered. Concentration in vacuo afforded 83 mg crude material which was purified via silica gel to give 11 mg(16%) of 3. Maldi MS : M + H = 560.4. (MW = 560.79) . TLC (EtOAc) Rf = 0.08 (major diast.) & 0.16 (minor diast.) . TLC (5% MeOH/EtOAc) Rf = 0.18 (major diast.) & 0.26 (minor diast.) . HPLC retention time was 11.05 min.
""H NMR (CDC13) d 8.50 (m, 2H) , 7.35-7.02 (m, 7H) , 5.89 (m, IH), 4.05-3.78 (m, 3H) , 3.37-2.69 (m, 5H) , 2.62- 2.45 (m, 4H) , 2.26 ( , 2H) , 2.08-1.16 (m, 15H) , 1.38 (s,9H) .
Example 63
Synthesis nf Compound 130
Figure imgf000239_0001
In an oven-dried 25 L round-bottomed flask, alkyne lactam 1 (54.6 mg, 0.682 mmol) was dissolved in 5 mL of DMF. Sodium hydride (34.4 mg of a 60% dispersion in mineral oil, 0.860 mmol) was added with cooling using an ice-bath. Gas evolution was observed. (S)- Epichlorohydrin (60 uL, 0.765 mmol) was added. The mixture was stirred overnight at room temperature, then the decahydroisoquinoline amide (182 mg, 0.770 mmol) was added. The mixture was heated to 80 °C overnight. The mixture was cooled, poored into water, and extracted with CH2Cl2. The organic extract was washed several times with water, dried (MgS04), and evaporated in vacuo to afford a yellow residue that was punfiec by preparative HPLC to afford the a diastereomeric mixture of alkyne DHIQ lactam 2 (120 mg, 34%) as a light yellow oil. HPLC: retention times of 13.57, 13.67, 13.87 minutes in a 5:1:1 ratio respectively. H NMR : d 1.3-1.4 three singlets m a 2:1:1 ratio; 1.4- 2.7 (several overlapping multiplets, 2.8-2.95 (multiplet), 3.0-3.7 (multiplet) , 3.8-4.1 (multiplet), 5.95-6.05 (multiplet), 6.1, 6.18, 6.32, 6.4 (broad singlets in a 1:1:1:1 ratio), 6.2-6.3 (doublet of doublets), 7.15-7.35 (multiplet). MALDI-MS: peak at 506.3 (M + H+) .
Example 64
Synthesis of Compound 124
Figure imgf000240_0001
THF, -7B°C to r.t
The lactam 1 (0.13 mmol) was dissolved in dry THF at -78 °C and to this solution was added Phosphazene Base P4-t-Bu (Fluka, 1.0M in hexane, 0.14 mmol). After stirring 15 minutes the anion solution was added via cannula to a solution of epoxide 2 (0.13 mmol) dissolved in dry THF at -78 °C and the reaction was allowed to warm to room temperature and stir overnight. The reaction was then diluted with water and extracted with ethyl acetate. The organic layer was washed sequentially with saturated aqueous NaHC03 and brine, followed by drying (MgS04), filtration and concentration in vacuo. The crude residue was taken up in dichloromethane and filtered through a plug of silica gel eluting with 8% MeOH in dichloromethane. Product containing fractions were concentrated in vacuo and the resultant residue further purified by preparative HPLC (column: Delta-Pak C18 15mm lOOA 19x300 mm. Gradient: 20% to 100% acetonitrile in water with 0.1% TFA. Flow rate: 20 ml/min. Detection: 214 nm) to yield 3 mg of product 3 as a mixture of diastereomers; TLC Rf = 0.44 (8% MeOH/CH2Cl2) ; HPLC Rt = 14.8, 14.9 min (95%); MALDI- TOF MS m/z 561 (M+) ; 1H NMR (CDC13) d 7.35-7.10 (m, 7 H) , 6.73 ( , 1 H) , 6.58 (d, 2 H) , 5.82 (br s, 1 H) , 4.12-3.85 (m, 4 H) , 3.51 (m, IH) , 3.30 (m, IH) , 2.92
(m, 1 H) , 3.63-2.20 (m, 4 H) , 2.05-1.12 (m, 18 H) , 1.38 (s, 9 H) .
Example 65
Synthesis of Compound 127
Phosphazene Base P4-t-Bu
Figure imgf000242_0002
THF -78°C o r t
Cyanomethyl lactam 1 (82 mg, 0.38 mmol) was dissolved m anhydrous THF (2 L) and cooled to -78 °C. Phosphazene Base P4-t-Bu (Fluka, 1.OM in hexane, 380 uL, 0.38 mmol) was added to give a yellow anion. The anion solution was stirred at -78 °C for 35 minutes and was then cannulated under nitrogen over 30 seconds into a -78 °C solution of 2 (112 mg, 0.38 mmol) in 2mL of THF and was washed m with 0.5 mL of THF. The reaction was gradually warmed to room temperature over 4 hr, then stirred at room temperature for 3 days. The reaction was cooled to -78 °C, quenched with 0.5 mL of a saturated ammonium chloride solution, and concentrated m vacuo to remove the THF. The residue was then partitioned between ethyl acetate and saturated bicarbonate solution and the aqueous layer was extracted with ethyl acetate. The combined organic layers were then washed with water, brme and dried (MgS04) and filtered. Concentration m vacuo afforded 375 mg crude material which was purified via silica gel (8 : 2, ethyl acetate: CH2C12, to give 118 mg(61%) of 3 that was < 80% pure. 58 mg was purified via prep. HPLC to give 10 mg of pure material as a 2: 1 mixture of diastereomers. HPLC retention times were 12.73 mm. (67%) & 12.86 mm. (33%) . Maldi MS: M + H = 510.47 (MW = 508.71) . TLC ( EtOAc) Rf = 0.37 & 0.31. 1H NMR (CDC13) d 7.38-7.13 (m, 5H) , 6.09-5.82 (br s, IH) , 4.29-3.96 (m, 3H) , 3.84 (m, IH) , 3.49-2.91 (m, 5H) , 2.77-2.18 (m, 9H) , 2.10-1.20(m, 11H) , 1.39 (s,9H) .
Example 66
Synthesis of Compound 131
Figure imgf000243_0001
THF -78°C to r t
The lactam 1 (0.061 mmol) was dissolved in dry THF at -78 °C and to this solution was added Phosphazene Base P4-t-Bu (Fluka, 1.0M in hexane, 0.067 mmol) . After stirring 15 minutes the anion solution was added via cannula to a solution of epoxide 2 (0.061 mmol) dissolved in dry THF at -78 °C and the reaction was allowed to warm to room temperature and stir overnight. The reaction was then diluted with water and extracted with ethyl acetate. The organic layer was washed sequentially with saturated aqueous NaHC03 and brine, followed by drying (MgS04), filtration and concentration in vacuo . The crude residue was purified by flash silica gel chromatography eluting with 3% MeOH in dichloromethane to give 2.1 mg of product 3 as a 1:1 mixture of diastereomers; TLC R^ = 0.14 (2:1 ethyl acetate/hexanes); HPLC Rt = 13.6, 13.8 min (68%); MALDI-TOF MS m/z 580 (M+) ; 1H NMR (CDC13) d 7.32-7.08 (m, 5 H) , 5.86 (br s, 1 H) , 4.08-3.73 (m, 4 H) , 3.65- 3.14 (m, 4H) , 3.00-2.49 (m, 8H) , 2.41-0.92 (m, 13 H) , 2.27 (s, 1.5 H) , 2.22 (s, 1.5 H) , 2.16 (s, 1.5 H) , 2.11 (s, 1.5 H) , 1.46 (s, 9 H) .
Example A
Synthesis of Compound 126
Figure imgf000244_0001
THF, -7θ°C to r.t
The lactam 1 (0.20 mmol) was dissolved in dry THF at -78 °C and to this solution was added Phosphazene Base P4-t-Bu (Fluka, 1.0M in hexane, 0.21 mmol) . After stirring 15 minutes the anion solution was added via cannula to a solution of epoxide 2 (0.20 mmol) dissolved in dry THF at -78 °C and the reaction was allowed to warm to room temperature and stir overnight. The reaction was then diluted with water and extracted with ethyl acetate. The organic layer was washed sequentially with saturated aqueous NaHC03 and brme, followed by drying (MgS04) , filtration and concentration in vacuo. The crude residue was taken up in dichloromethane and filtered through a plug of silica gel eluting with 3% MeOH in dichloromethane. Product containing fractions were concentrated in vacuo and the resultant residue further purified by preparative HPLC (column: Delta-Pak Cι_s 15mm lOOA 19x300 mm. Gradient: 20% to 100% acetonitrile in water with 0.1% TFA. Flow rate: 20 ml/mm. Detection: 214 nm) to yield 2.5 mg of product 3 as a mixture of diastereomers; TLC = 0.21 (3% MeOH/CH2Cl2) ; HPLC Rt = 14.8 min (98%); MALDI-TOF MS m/z 588 (M+) .
Figure imgf000245_0001
In an oven-dried 25 mL round-bottomed flask, isoxazole lactam 1 (54.6 mg, 0.201 mmol) was dissolved m 3 mL of THF. (S) -Epichlorohydrin (20 uL, 0.255 mmol) was added. P-4-tBu phosphazene base (210 uL, 0.210 mmol) was added dropwise via syringe initially producing a dark orange-brown color that faded. The mixture was stirred for 30 minutes at room temperature and the mixture was poured into water and extracted with CH2C12. The organic extract was dried (Na2S04) and evaporated in vacuo.. The residue was dissolved in anhydrous CH3CN and the decahydroisoquinoline amide (54.4 mg, 0.230 mmol) was added. The mixture was refluxed overnight. The solvent was evaporated and the residue was purified by preparative HPLC to afford the isoxazole DHIQ lactam 2 (38 g, 34%) as a light yellow oil. HPLC: retention times of 12.28, 12.86, 13.68 minutes at 93% purity. 1H NMR : d 1.3-1.4 three singlets in a 4:4:1 ratio; 1.4-2.7 (several overlapping multiplets, 1.4-2.3 (several overlapping multiplets), 2.45-3.35 (several overlapping multiplets), 3.35-4.1 (several multiplets), 4.3-4.4 (doublet), 5.8 (multiplet), 5.9, 6.0, and 6.3 (three broad singlets in a ratio of 4:4:1 ratio), 7.1-7.2 (multiplet), 7.2-7.4 (multiplet) . MALDI-MS: calc'd: 564.9; found 565.5 (M + H+).
Example §9
Synthesis of Compound 125
Figure imgf000247_0001
THF,-78°Ctor.t.
The lactam 1 (0.12 mmol) was dissolved in dry THF at -78 °C and to this solution was added Phosphazene Base P4-t-Bu (Fluka, 1. OM in hexane, 0.13 mmol) . After stirring 15 minutes the anion solution was added via cannula to a solution of epoxide 2 (0.12 mmol) dissolved in dry THF at -78 °C and the reaction was allowed to warm to room temperature and stir overnight. The reaction was then diluted with water and extracted with ethyl acetate. The organic layer was washed sequentially with saturated aqueous NaHC03 and brine, followed by drying (MgS04), filtration and concentration in vacuo . The crude residue was taken up in dichloromethane and filtered through a plug of silica gel eluting with 3% MeOH in dichloromethane. Product containing fractions were concentrated in vacuo and the resultant residue further purified by preparative HPLC (column: Delta-Pak C18 15mm lOOA 19x300 mm. Gradient: 20% to 100% acetonitrile in water with 0.1% TFA. Flow rate: 20 ml/min. Detection: 214 nm) to yield 1.5 mg of product 3 as a single diastereomer; TLC Rf = 0.27 (3% MeOH/CH2Cl2)
HPLC Rt = 14.7 min (100' MALDI-TOF MS m/z 576
(M+) ; 1H NMR (CDC13) d 7.40-7.15 (m, 7 H) , 6.70 (m, 1 H), 6.55 (d, 2 H) , 5.80 (br s, 1 H) , 4.28 (m, 1 H) , 4.05-3.90 (m, 2 H) , 3.70-3.38 (m, 2H) , 3.20 (m, IH) , 3.00-2.75 (m, 2 H) , 2.70 (s, 3 H) , 2.55 (m, 2H) , 2.30
(m, 2H) , 2.20-0.80 (m, 14 H) , 1.35 (s, 9 H) .
Example 70
Sunthesis of Compound 128
Figure imgf000248_0001
EtOOC jl
Figure imgf000248_0002
The lactam 1 (90 mg, 0.28 mmol) was dissolved in THF (3 mL) and cooled to -78 CC. This was followed by the addition of the phosphazene base (Fluka; IM in hexane, 0.28 mL, 0.28 mmol). After stirring at -78 °C for one hour the epoxide was added as a solution in 1 mL THF. The reaction was then warmed to 25 °C and stirred for an additional 3 hours. The reaction was then quenched by the addition of water and extracted by ethyl acetate. The organic portion was then dried over MgS04, filtered and concentrated in vacuo. The crude oil was purified by silica gel chromatography, eluting with 1:1, ethyl acetate:hexanes, this provided the two major products (HPLC. indicated two components for each isolate) .
B.
Figure imgf000249_0001
To the elaborated lactam 1 (40 mg) in 2:1, THF:H20 (5 mL) was added LiOH (2 eq.) . The reaction was then stirred at 40 °C for 16 hours. TLC indicated the formation of a new component. The reaction was diluted by ethyl acetate, after which the organic portion was separated, dried over MgS04, filtered and concentrated in vacuo . to yield product 2 as a mixture of diastereomers .
1H NMR (CDC13) : d 7.10-7.50 ( , 10 H) , 5.90-6.15 (m, IH), 3.90-4.40 (m, 2H) , 3.20-3.70 (m, 3H) , 2.80-3.10 ( , 2H) , 2.60-2.70 (m, 2H) , 2.20-2.60 ( , 3H) , 1.60- 2.10 (m, 9H), 1.40 (q, 15H) , 1.20-1.40 (m, 8H) . Example 71
Synthesis of Compound 259
Figure imgf000250_0001
Phosphazene Base Rj-t-Bu
Figure imgf000250_0002
THF, -78°Ctort
The lactam 1 (0.11 mmol) was dissolved in dry THF at -78 CC and to this solution was added Phosphazene Base P4-t-Bu (Fluka, 1.OM in hexane, 0.12 mmol). After stirring 15 minutes the anion solution was added via cannula to a solution of epoxide 2 (0.11 mmol) dissolved in dry THF at -78 °C and the reaction was allowed to warm to room temperature and stir overnight. The reaction was then diluted with water and extracted with ethyl acetate. The organic layer was washed sequentially with saturated aqueous NaHC03 and brme, followed by drying (MgS04), filtration and concentration in vacuo . The crude residue was taken up in dichloromethane and filtered through a plug of silica gel eluting with 5% MeOH in dichloromethane. Product containing fractions were concentrated in vacuo and the resultant residue further purified by preparative HPLC (column: Delta-Pak C18 15mm lOOA 19x300 mm. Gradient: 20% to 100% acetonitrile in water with 0.1% TFA. Flow rate: 20 ml/mm. Detection: 214 nm) to yield 12 mg of product 3; TLC Rf = 0.50 (8% MeOH/CH2Cl2) ; HPLC Rt = 12.8 min (100%) MALDI-TOF MS m/z 541 (M+) ; XH NMR (CDC13) d 7.35-7.16
(m, 5 H) , 5.86 (br s, 1 H) , 4.08-3.76 ( , 4 H) , 3.49- 3.22 ( , 4 H), 2.89 (br s, 1 H) , 2.50 (m, 2 H) , 2.25
(br s, IH) , 2.14-1.11 (m, 22 H) , 1.38 (s, 9 H) .
Exampl 72
Synthesis of Compound 260
Figure imgf000251_0002
Figure imgf000251_0001
In an oven-dried 25 mL round-bottomed flask, triazole lactam 1 (124 mg, 0.358 mmol) was. dissolved in 5 mL of THF. (S) -Epichlorohydrin (50 uL, 0.639 mmol) was added. P-4-tBu phosphazene base (370 uL of 1.0M solution in hexane, 0.370 mmol) was added dropwise via syringe initially producing a dark orange-brown color that faded. The mixture was stirred for 30 minutes at room temperature and the decahydroisoquinoline amide (124 mg, 0.525 mmol) was added. The mixture was refluxed overnight. The solvent was evaporated and the residue was purified by preparative HPLC to afford the triazole DHIQ lactam (189.1 mg, 84%) . HPLC: retention times of 12.94, 14.22 minutes at 99% purity. 1H NMR : dl.3-1.4 two singlets in a 1:1 ratio; 1.4-3.1 (several overlapping multiplets, 1.4-2.3 (several overlapping multiplets), 2.45-3.35 (several overlapping multiplets), 3.2-4.2 (several multiplets), 5.4-5.6 (multiplet), 6.1 and 6.45 (two broad singlets in a 1:1 ratio), 5.9, 6.0, and 6.3 (three broad singlets in a ratio of 4:4:1 ratio), 7.1 (doublet), 7.2-7.5 (multiplet) . MALDI-MS: calc'd (-DHIQ) : 401.2; found 403.6 (M - DHIQ + 2H+) .
Example 73
Synthesis of Compound 129
Figure imgf000252_0001
In a heavy-walled screw-top test tube, the alkyne lactam 1 (83 mg, 0.164 mmol) was dissolved in 5 mL of xylene. Tributyltin azide (200 mg, 0.602 mmol) was added, the tube was sealed and heated to 205° C overnight. The dark brown solution was cooled and directly chromatographed using a gradient from CH2C12 to 50% EtOAc/MeOH to afford the triazole product 2 (14 mg, 2.5%) as a light yellow oil. HPLC: retention times of 12.01, 12.44, 13.01, 13.22 minutes in an 8:4:1:1 ratio at 99% purity. MALDI-MS: calc'd (-DHIQ) : 550.4; found 552.9 (M + 2H+) . Example 4
Synthes is of Compound 227
Figure imgf000253_0001
3 ) isopropanol conc HCl rt
To a cooled solution (-78 °C) of lactam 1 (0.10g, 0.46mmol) in anhydrous THF (l.OmL) was added phosphazene base P4 t-butyl solution (1.0M in hexanes, 0.46mL, 0.46mmol) with stirring. After a 15 mm. stirring period, epoxide 2 (0.173g, 0.46mmol) was added in one portion and the reaction was allowed to slowly warm to rt. After 0.5h at rt, 1. OM HCl (lO.OmL) was added and the solution was diluted with ethyl acetate (60mL) . The ethyl acetate was washed with sat. NaHC03 (1 x lOmL), brme (1 x lOmL) dried (MgS04), filtered, and evaporated to give a brown foam. The crude acetonide (0.270g, 0.46mmol) was dissolved isopropanol (lOmL) and treated with cone. HCl (3.0mL) at rt. After 2.Oh., the solution was adjusted to pH 11 with 3.ON NaOH and extracted with ethyl acetate (3 x75mL) . The ethyl acetate was dried (MgS04) and evaporated to give the crude product which was purified by column chromatography: methylene chlonde/methanol (98/2) to give the product as an off white solid (0.090g, 36%) . MS: crude acetonide: M+Na = 617; product: M+Na = 577 λE NMR (CDC13) 0.90 (m, 6H) ; 1.15(m, IH) ; 1.40 (m, IH) ; 1.50-1.80 ( , 2H) ; 1.90 (m, IH) ; 2.18 (m, 2.25H); 2.30-2.50(m, IH) ; 2.60 (m, 0.75H); 2.80-3.10(m, 4H) ; 3.30 (m, 2H) ; 3.60(m, 1.25H); 3.80 (m, 1.75H) ; 3.95(m, IH) ; 4.25(m, IH) ; 4.40 (m, 0.75H); 5.00(m, 0.25H); 5.25 ( , IH) ; 5.95(d, 0.25H); 6.10(d, 0.75H) ; 7.00-7.40(m, 14H)
Example 75
Synthes i s of Compound 232
Figure imgf000254_0001
Prepared using the procedure outlined in Example 24. The acetonide was purified by column chromatography: 60/40 hexane/ethyl acetate. MS: M+NA = 647. The product was purified by column chromatography: 98/2 CH2Cl2/MeOH. MS: M+H = 585 XH NMR (CDCI3) 1.70 (m, 2H) ; 1.80 ( , IH) ; 1.90 (m, IH) ; 2.10(m, IH) ; 2.40-3.10(m, 10H) ; 3.60(s, 3H) ; 3.75 (m, IH) ; 3.90 (m, IH) ; 4.0 (m, IH) ; 4.30 (m, 3H) ; 5.30(m, IH) ; 6.10(d, IH) ; 7.00- 7.40(m, 14H) . Example 76
Synthesis of Compound 231
Figure imgf000255_0001
Prepared using the procedure outlined in Example 24, The acetonide was purified by column chromatography: 98/2 CH2Cl2/MeOH. MS: M+H = 642. The product was purified by column chromatography: 96/4 CH2Cl /MeOH. MS: M+H = 602 XH NMR (CDC13) 1.50-2.50 (m, 6H) ; 2.50- 3.40 ( , 6H) ; 3.50-4.40(m, 7H) ; 5.25(m, IH) ; 5.95(m, IH) ; 7.00-7.60(m, 18H) .
Example 77
Synthesis of Compound 216
cone. HCl
Figure imgf000255_0002
Figure imgf000255_0003
Prepared using the procedure outlined in Example 24. The acetonide was purified by column chromatography: 50/50 hexane/ethyl acetate. MS: M+NA = 645. The product was purified by column chromatography: 96/4 CH2Cl2/MeOH. MS: M+NA = 605 1H NMR (CDC13) 1.10-1.40 ( , 2H) ; 1.70 (m, 2H) ; 1.80-2.10 ( , 4H) ; 2.35 (m, 0.5H) ; 2.50 (m, IH) ; 2.65 (m, 0.5H) ; 2.80-3.10 (m, 4H) ; 3.20 (m, 2H) ; 3.30-3.55 (m, 3H) ; 3.70 (m, IH) ; 3.80-4.00 (m, 4H) ; 4.25(m, IH) ; 4.37 (m, IH) ; 5.27(m, IH) ; 6.15(d, IH) ; 7.10-7.40(m, 14H) .
Example 78
Synthesis of Compound 221
Figure imgf000256_0001
Figure imgf000256_0002
Figure imgf000256_0003
Prepared using the procedure outlined in Example 24. The acetonide was not purified by column chromatography. MS: (crude) M+H = 644. The product was purified by column chromatography: 96/4 CH2Cl2/MeOH. MS: M+H = 604 1H NMR (CDC13) 1.40-2.20 (m, 6H) ; 2.30 (m, IH) ; 2.50-3.40(m, 9H) ; 3.75(m, 2H) ; 4.00(m,lH) ; 4.25(m, IH) ; 5.30(m, IH) ; 6.35(d, 0.5H) ; 6.50(d, 0.5H) ; 7.00- 7.40 (m, 14H) ; 7.50 (m, 2H) ; 8.50 (m, 2H) . Example
Synthesis of Compound 223
Figure imgf000257_0001
Prepared using the procedure outlined in Example 24. The acetonide was not purified by column chromatography. MS: (crude) M+NA = 670. The product was purified by column chromatography: 97/3 CH2Cl /MeOH. MS: M+NA = 630 λE NMR (CDC13) 1.40 (m, IH) ; 1.30-1.80 (m, 2H); 1.95 (m, IH) ; 2.10(m, IH) ; 2.25 ( , 2H) ; 2.30- 3.40 (m, 7H) ; 3.60-3.80(m, 2H) ; 3.85(m,lH); 4.00 (m, IH) ; 4.25(m, IH) ; 4.45(m,lH); 5.30(m, IH) ; 5.80(m,lH) ; 6.15(d, IH) ; 7.10-7.40(m, 14H) .
Example 80
Synthesis of Compound 230
Figure imgf000258_0001
Prepared using the procedure outlined in Example 24. The acetonide was not purified by column chromatography. MS: (crude) M+NA = 746. The product was purified by column chromatography: 97/3 CH2Cl2/MeOH. MS: M+NA = 706.
Example 81
Synthesis of Compound 224
Figure imgf000258_0002
Prepared using the procedure outlined in Example 24. The acetonide was not purified by column chromatography. MS: (crude) M+NA = 673. The producr. was purified by column chromatography: 96/4 CH2Cl2/MeOH. MS: M+NA = 633 "'H NMR (CDC13) 0.090-1.30 ( , 4H) ; 1.40- 1.80(m, 4H) ; 1.90-2.35 (m, 3H) ; 2.45 ( , IH) ; 2.65 (m, IH) ; 2.70-3.10(m, 6H) ; 3.25 (m, 3H) ; 3.60-4.00(m, 6H) ; 4.25 (m, IH) ; 4.35 (m, 0.5H); 4.75(m, 0.5H) ; 5.25 (m, IH) ; 6.20 (m, IH) ; 7.10-7.40(m, 14H) .
E &mple 82
Synthesi s of Compound 225
Figure imgf000259_0001
3.) isopropanol conc. HCl rt
Prepared using the procedure outlined in Example 74. The acetonide was not purified by column chromatography. MS: (crude) 2M+NA = 1179. The product was purified by column chromatography: 80/20 ethyl acetate/hexane. MS: M+H = 539 1H NMR (CDC13) 0. 55 (m, IH) ; 0.659m, IH) ; 0.95 (m, IH) ; 1.05(m, IH) ; 1.75 (m, 2H) ; 1.95 (m, IH) ; 2.20 (dd, IH) ; 2.65 (dd, IH) ; 2.70- 3.10 ( , 6H) ; 3.20(d, IH) ; 3.65(dd, IH) ; 3.95m, 2H) ; 4.25(t, IH) ; 5.25(m, IH) ; 5.95(d, IH) ; 7.10-7.40(m, 14H) . Example 8
Synthesis of Compound 226
Figure imgf000260_0001
Prepared using the procedure outlined in Example 24.
The acetonide was purified by column chromatography: 60/40 hexane/ethyl acetate. MS: M+NA = 666. The product was purified by column chromatography: 40/60 hexane/ethyl acetate. MS: M+H = 604 1H NMR 1. 55 (m, 0.5H) ; 1.70m, 0.5H); 1.95(m, IH) ; 2.50 ( , IH) ; 2.70-3.10(m, 7.5H); 3.15(dd, IH) ; 3.30(m, IH) ; 3.40(m, IH) ; 3.75 (m, IH) ; 3.80-4.10 ( , 2H) ; 4.25(m, 2.5H); 4.45(m, 0.5H) ; 5.25 (m, IH) ; 6.15(m, IH) ; 6.45(d, IH) ; 6.55(q, IH) ; 6.70(q, IH) ; 7.10-7.40(m, 16H) .
Example 9
Synthesis of Compound 229
Figure imgf000261_0001
Prepared using the procedure outlined in Example 74 The acetonide was purified by column chromatography and the diastenomers were isolated separately. MS: (isomer 1) M+NA = 642; (isomer 2) M+NA = 642. The individual diastereomers were deprotected and purified by column chromatography: 98/2 CH2Cl2/MeOH to give isomer 1 and isomer 2. MS: (isomer 1) M+NA = 602; (isomer 2) M+NA = 602. λU NMR (CDC13) isomer 1: 1.05 (d, IH) ; 1.35(s, 3H) ; 1.45 (s, 3H) ; 1.75 (m, IH) ; 1.90-2.20 ( , 3H) ;
2.65(m, IH) ; 2.70-3.10 (m, 8H) ; 3.70 m, IH) ; 3.95 (m,
2H) ; 4.20(m, IH) ; 4.35 (m, IH) ; 5.25(m, IH) ; 6.05(d,
IH) ; 7.10-7.40(m, 14H) . λR NMR (CDCI3) isomer 2: 1.10
(d, IH) ; 1.40 (s, 3H) ; 1.50 (s, 3H) ; 1.75(m, IH) ;
1.95 (m, IH) ; 2.15(m, IH) ; 2.50(m, 2H) ; 2.80-3.10 (m,
6H) ; 3.35(m, 2H) ; 3.65 (m, IH) ; 3.80 (m, IH) ; 4.00 (m,
2H) ; 4.25(m, IH) ; 5.25 (m, IH) ; 5.95(d, IH) ; 7.10- 7.40 (m, 14H) . Example 85
Synthesis of Compound 261
Figure imgf000262_0001
Prepared using the procedure outlined in Example 74. The acetonide was purified by column chromatography: 60/40 hexane/ethyl acetate and the diastenomers were isolated separately. MS: (isomer 1) M+H = 658; (isomer 2) M+H = 658. The individual diastereomers were deprotected and purified by column chromatography: 40/60 hexane/ethyl acetate to give isomer 1 and isomer 2. MS: (isomer 1) M+H = 618; (isomer 2) M+NA = 640. 1H NMR (CDC13) isomer 1: 1.75(m, IH) ; 1.90-2.20 { , 3H) ; 2.70(s, 3H) ; 2.75-3.15 (m, 6H) ; 3.75(m, IH) ; 4.00(m, 3H) ; 4.25(m, IH) ; 4.65 (m, IH) ; 5.25(m, IH) ; 6.05(d, IH) ; 6.55(dd, 2H) ; 6.70(m, IH) ; 7.00-7.40 (m, 16H) . XH NMR (CDCI3) isomer 2: 1.70 (m, 2H) ; 1.95 (m, IH) ; 2.25(m, IH) ; 2.55(m, IH) ; 2.70(s, 3H) ; 2.80-3.10(m, 8H) ; 3.35(dd, IH) ; 3.40(dd, IH) ; 3.75 (m, IH) ; 3.80 (m, IH) ; 4.05(m, IH) ; 4.25 (m, IH) ; 4.55(t, IH) ; 5.30(m, IH) ; 6.05(d, IH) ; 6.70(m, 2H) ; 7.10-7.40 (m, 17H) . Example 86
Synthesis of Compound 228
Figure imgf000263_0001
The benzyl triazole from Example 80 was purified (and diastereomers isolated) by column chromatography: 97/3 CH2Cl2/MeOH. MS: M+NA = 706. The individual benzyl protected diastereomers were dissolved in MeOH and combined with 20% Pd/C (cat.) . Each solution was hydrogenated under pressure (50 psi) at rt for 5 days and the resulting crude product was purified by column chromatography 96/4 CH2Cl2/MeOH to give isomers 1 and 2. MS: (isomer 1) M+H = 594; (isomer 2) M+NA = 616. 1H NMR (CDC13) isomer 1: 1.60 (m, IH) ; 1.80 (m, 2H) ; 2.40 (s, IH) ; 2.60-3.15( , 10H) ; 3.65(m, IH) ; 3.80(m, IH) ; 4.00(m, IH) ; 4.20(m, IH) ; 5.25 (m, IH) ; 6.90(m, IH) ; 7.00-7.40 (m, 14H) . *H NMR (CDC13) isomer 2: 1.30 (m, IH) ; 1.75(m, IH) ; 1.95 (m, 2H) ; 2.35(m, IH) ; 2.50 (m, IH) ; 2.80-3.10(m, 8H) ; 3.25(d, IH) ; 3.65(m, IH) ; 3.80(m, IH) ; 4.05(m, IH) ; 4.30 (m, IH) ; 4.50(m, IH) ; 5.25(m, IH) ; 6.75 (m, IH) ; 7.10-7.40(m, 14H) . Example 87
Synthesis of Compound 219
Figure imgf000264_0001
separated diastereomers
Figure imgf000264_0002
Isomer 1: Prepared using the procedure outlined in Example 24. The acetonide was purified by column chromatography 30/70 hexane/ethyl acetate MS: M+NA =
645. The product was purified by column chromatography: 30/70 hexane/ethyl acetate MS: M+NA = 605 1H NMR (CDC13) 1.45(m, IH) ; 1.70 (m, IH) ; 1.80-2.05 (m, 4H) ; 2.25 (q, IH) ; 2.35(q, IH) ; 2.65(m, IH) ; 2.75-3.10 (m, 8H) ; 3.60 ( , 3H) ; 3.75(m,lH); 3.85 (m, IH) ; 3.95(m, 2H) ; 4.25(m, IH) ; 5.25(m, IH) ; 6.05(m, IH) ; 7.10-7. 0 (m, 14H) .
Isomers 2,3: (Chiral center with THF ring has opposite configuration to that of isomer 1 above) . Prepared using the procedure outlined m Example 74. The acetonide was purified (diastereomers isolated) by column chromatography 30/70 hexane/ethyl acetate MS: M+NA = 645. The individual diastereomers were purified by column chromatography: 30/70 hexane/ethyl acetate MS: (isomer 2) M+NA = 605 (isomer 3) M+NA = 605 XH NMR (CDCI3) (isomer 2) 1.45 (m, 2H) ; 1.90 ( , 2H) ; 2.10 (m, IH) ; 2.30 (m, IH) ; 2.35 (m, IH) ; 2.45 (m, IH) ; 2.75- 3.10(m, 6H) ; 3.25(m, 2H) ; 3.65(m, 3H) ; 3.75(m, 3H) ; 3.95 ( , 2H) ; 4.25(m, 2H) ; 5.25(m, IH) ; 6.10(m, IH) ; 7.10-7.40 (m, 14H) . 1H NMR (CDC13) (isomer 3) 1.15 ( , IH) ; 1.80 (m, IH) ; 1.95(m, 2H) ; 2.10(m, IH) ; 2.25 ( , IH) ; 2.40 ( , IH) ; 2.60(m, IH) ; 2.75-3.10(m, 8H) ;
3.40(m, IH) ; 3.60-4.00(m, 6H) ; 4.25(m, IH) ; 5.25(m, IH) ; 6.05(m, IH) ; 7.10-7.40 ( , 14H) .
Example 88
Synthesis of Compound 233
Figure imgf000265_0001
Prepared using the procedure outlined in Example 74. The acetonide was purified (diastereomers isolated) by column chromatography 45/55 hexane/ethyl acetate MS:
M+NA = 692. The individual diastereomers were purified by column chromatography: 35/65 hexane/ethyl acetate MS: (isomer 1) M+H = 630 (isomer 2) M+H = 630 1H NMR (CDCI3) (isomer 1) 1.75 (m, IH) ; 1.95 (m, IH) ; 2.10 (m, 2H) ; 2.75-3.10(m, 8H) ; 3.15(d, 2H) ; 3.30 (m, 2H) ;
3.80(m,2H); 4.00 ( , 2H) ; 4.25 ( , IH) ; 5.25(m, IH) ; 6.00 (m, IH) ; 6.20(d, IH) ; 6.60(t, IH) ; 6.95-7.40(m, 18H) . λH NMR (CDCI3) (isomer 2) 1.75(m, IH) ; 1.95 (m, 2H) ; 2.15(m, IH) ; 2.55 (m, IH) ; 2.75-3.10(m, 8H) ; 3.20- 3.50 (m, 4H) ; 3.75 (m, IH) ; 3.85 (m, IH) 4.00 ( , IH) ;
4,25(m, IH) ; 4.35(m, IH) ; 5.25 (m, IH) ; 6.10 (m, IH)
6.30(m, IH) ; 6.56(t, IH) ;7.10-7.40 (m, 18 H) .
Example 89
Synthesis of Compound 234
Figure imgf000266_0001
3.) isopropanol/conc. HCl rt
Prepared using the procedure outlined in Example 74. The acetonide was purified by column chromatography: 97/3 CH2Cl2/MeOH. MS: M+H = 633. The product was not purified. MS: M+H = 593.
F.xamol e 90
Synthesi s o f Compound 235
Figure imgf000267_0001
A mixture of the trans isomer of the lactam above (0.125 g, 0.27 mmol) and 60% sodium hydride (0.010 g, 0.25 mmol) in dimethylformamide (4 mL) was stirred under a nitrogen atmosphere for 30 mm. The epoxide (0.113 g, 0.30 mmol) was added and the mixture was heated at 60 °C for 4h. The mixture was re-charged with 60% sodium hydride (0.015 g, 0.37 mmol) heated at 60 °C for an additional 1.5h, and stirred at ambient temperature for 18h. The mixture was diluted with dichloromethane, washed with br e, dried (MgS04), and concentrated m vacuo . The residue was purified by chromatography on silica gel, elutmg with hexane:ethyl acetate (7:3) then with hexane:ethyl acetate (1:1) to give O.llg (48%) of product as a brown oil. MS: AP+, 862 (M+Na) and AP-, 874 (M+Cl) .
B. A solution of the acetonide (O.llg, 0.13 mmol) in 2- propanol (7 mL) and concentrated hydrochloric acid (3 mL) was stirred at ambient temperature for 3h, neutralized with 2N sodium hydroxide, and extracted with diethyl ether. The extracts were dried (MgS0 ), filtered, and concentrated in vacuo to give 0.040 g (58%) yield of the crude product, which was used without further purification. MS: ES+, 550 (M+Na) and ES-, 562 (M+Cl) .
C.
A solution of the amme (0.14 g, 0.27 mmol), methylchloroformate (0.023 mL, 0.30 mmol) and Et3N (0.05 mL, 0.36 mmol) in dichloromethane (2 mL) was stirrred at ambient temperature under a nitrogen atmosphere for 18h. The volatiles were removed m vacuo, and the residue was purified by reverse phase preparative HPLC to give a tan oil. Lyophilization gave 0.012 g (8%) of the product as a white solid. MS: ES+,608 (M+Na). 1H NMR (CDC13) 1.71 ( , IH) ; 1.96 (m, IH) ; 2.11 (m, IH) ; 2.26 (m, IH) ; 2.71-3.05 (m, 8H) ; 3.50 (s, 3H) ; 3.65 (m, IH) ; 3.82 (m, IH) ; 4.00-4.39 (m, 5H) ; 5.28 (m, IH) ; 5.43 (s, IH) ; 6.40 (d, IH) ; 7.08-7.33 ( , 14H) .
Example 91
Synthesis of Compound 239
Figure imgf000269_0001
HCl rt
Prepared as described in Example 74 with the exception that water, rather than 1.0 N HCl was used to quench the reaction. MS (AP-) of the acetonide = 660 (M-l) . MS (AP+) of the product = 644 (M+Na) . 1HNMR of the product (CDC13) : d 1.68 (m, 3H) , 2.07 (m, 3H) , 2.54 (m, 2H) , 2.92 ( , 6H) , 3.43 (m, IH) , 3.78 (m, IH) , 4.00 (m, 2H) , 4.50 (m, IH) , 5.34 (m, IH) , 6.10 (m, IH) , 6.70 (m, IH), 7.24 ( , 18H)
Example 92
Synthesis of Compound 238
Figure imgf000269_0002
3.) isopropanol/coπc. HCl rt Prepared as described in Example 74 with the exception that water, rather than 1.0 N HCl was used to quench the reaction. MS (AP+) of the acetonide = 684 (M+Na) . MS (AP+) of the product = 644 (M+Na) . 1HNMR of the product (CDC13) : d 1.62 (m, 3H) , 2.00 (m, 3H) , 2.50 (m, 2H) , 2.80 (m, 6H) , 3.30 (m, 2H) , 4.00 (m, 2H) , 4.34 (m, IH) , 5.33 (m, IH) , 6.14 (m, IH) , 6.30 (m, IH) , 7.24 (m, 18H)
Example 93
Synthesis of Compound 240
Figure imgf000270_0001
3 ) isopropanol/conc HCl rt
Prepared as described in Example 74 with the exception that water, rather than 1.0 N HCl was used to quench the reaction. MS (AP+) of the acetonide = 691 (M+Na) . MS (AP+) of the product = 651 (M+Na) . ''HNMR of the product (CDCI3) : d 1.66 ( , 3H) , 2.08 (m, 3H) , 2.59 (m, 2H) , 2.95 (m, 6H) , 3.40 ( , IH) , 3.85 (m, IH) , 4.14 (m, 2H), 4.27 (m, IH) , 5.32 (m, IH) , 6.22 ( , IH) , 6.73 (m, IH) , 7.25 (m, 18H) . Example 94
Synthesis of Compound 241
Figure imgf000271_0001
3.) isopropanol/conc HCl rt
Prepared as described in Example 74 with the exception that water, rather than 1.0 N HCl was used to quench the reaction. MS (AP+) of the acetonide = 645 (M+l) . MS (AP+) of the product = 627 (M+Na) . 1HNMR of the product (CDC13) : d 1.70 (m, 3H) , 2.00 (m, 3H) , 2.58 (m, 2H), 2.97 (m, 6H) , 3.40 (m, IH) , 3.85 ( , IH) , 4.10 (m, 2H) , 4.32 (m, IH) , 5.33 (m, IH) , 6.30 (m, IH) , 6.80 ( , IH), 7.25 (m, 17H) , 8.01 ( , IH) .
Example 95
Synthesis of Compound 208
Figure imgf000272_0001
A .
The lactam (1.20 g, 2.69 mmol, 1 eq) was dissolved in anhydrous dimethylformamide (8 mL) under Argon and cooled with an isopropanol dry ice bath to -40 °C. A solution of sodium bis (trimethylsilyl) amide (1.0M THF, 2.69 mL, 2.69 mmol, 1 eq) was added dropwise via syringe and the reaction was stirred for 15 mm. maintaining the bath temp between -40 - -50 CC.
Dιhydro-5 (S) - [ [ [ (trifluoromethyl) sulfonyl] oxy]methyl] - 3 (R)- (phenylmethyl) -3 [ 2H) -furanone (J. Med. Chem., 1994, Vol. 37, No. 21, 3443-51; 1.00 g, 2.96 mmol, 1.1 eq) was added as a solid and the reaction was stirred vigorously for 10 mm. and then quenched with several drops of glacial acetic acid. The reaction mixture was evaporated in vacuo to a residue and partitioned between ethyl acetate, saturated aqueous brme, and water. After separating the layers, the aqueous layer was back-extracted with ethyl acetate. The combined organic layers were washed with saturated aqueous br e, dried over anhydrous magnesium sulfate, evaporated m vacuo and purified by flash silica gel chromatography eluting with ethyl acetate : hexane
(3:7) . Fractions containing the alkylated lactam were combined, evaporated in vacuo to provide 0.883 g (52 %) of product as a foam. MS (ESI) : M+Na = 657.
B. The butyrolactone (1.202 g, 1.89 mmol, 1 eq) from step A was dissolved at ambient temperature in dimethoxyethane (20 mL) and cooled with an ice water bath. Aqueous lithium hydroxide (1.0 N, 4.75 mL, 4.75 mmol, 2.5 eq) was added via pipette and the mixture was stirred for 0.5 h. The reaction was warmed to ambient temperature and stirred for an additional 1 h. Aqueous citric acid (10% w/v) was added to reach an acidic pH and the mixture was evaporated in vacuo. The residue was partitioned between ethyl acetate : diethyl ether (4:1) and aqueous citric acid (10% w/v) . After separating the layers, the aqueous layer was back- extracted with ethyl acetate. The combined organic layers were washed with water, saturated aqueous br e, dried over anhydrous magnesium sulfate, evaporated n vacuo and dried under high vacuum to provide the acid (1.32 g, 106%) as a foam. MS (APCI) : M - 1 = 651.
C.
The acid (1.28 g, 1.97 mmol, 1 eq) from step B m 5 L anhydrous dimethylformamide under Argon was combined with lmidizole (1.472 g, 21.6 mmol, 11 eq) followeα by tertbutyldi ethylsilyl chloride (2.96 g, 19.7 mmol, 10 eq) and stirred at ambient temperature for 16 h. Tne reaction was quench by addition of methanol (15 mL) and stirred for an additional 45 mm. Aqueous lithium hydroxide (1.0 N, 2.0 mL, 1 eq) was added and the mixture was evaporated in vacuo. The residue was partitioned between ethyl acetate and aqueous sodium hydrogen sulfate (1.0 N) . After separating the layers, the aqueous layer was back-extracted with ethyl acetate. The combined organic layers were washed with saturated aqueous brme, dried over anhydrous magnesium sulfate, evaporated m vacuo and dried under high vacuum to provide the silyl protected acid (1.46 g, 97%) as a foam. MS (APCI): M - 1 = 766.
D.
The silyl protected acid ( 1 . 32 g, 1 . 72 mmol , 1 eq) from step C in anhydrous dimethylformamide ( 7 mL ) under Argon was treated consecutively with dusopropylethyl amme ( 0 . 316 mL, 1 . 81 mmol , 1 . 05 eq) , 1- hydroxybenzotnazole (0.244 g, 1.81 mmol, 1.05 eq) , (1S,2R)- (-)-l-ammo-2-mdanol (0.283 g, 1.90 mmol, 1.1 eq) , and 1- (3-dιmethylamιnopropyl) -3-ethylcarbodιιmιde hydrochloride (0.347 g, 1.81 mmol, 1.05 eq) . After stirring at ambient temperature for 3 h, the reaction mixture was evaporated m vacuo, and partitioned between ethyl acetate, saturated aqueous brine, and water. After separating the layers, the aqueous layer was back-extracted with ethyl acetate. The combined organic layers were washed with saturated aqueous brine, dried over anhydrous magnesium sulfate, evaporated in vacuo and purified by flash silica gel chromatography eluting with ethyl acetate : hexane
(3:7). Fractions containing the product were combined, evaporated in vacuo and dried under high vacuum to provide the protected amide (1.06 g, 69%) as a foam. MS
(ESI) : M+Na = 920.
E.
The protected amide (1.035 g, 1.15 mmol, 1 eq) from step D was dissolved in trifluoroacetic acid (15 mL) and stirred under Argon for 15 min. The reaction was evaporated in vacuo and trituratated with diethyl ether/hexane. After decanting the mother liquor, the residual solid was dried under high vacuum to provide a partially deprotected product. The crude material was dissolved again in trifluoroacetic acid (15 mL) and stirred for 20 min. under Argon. The reaction mixture was evaporated in vacuo to a residue and triturated with hexane/diethyl ether. The slurry was filtered, washed with hexane and dried under high vacuum to provide the deprotected amine (0.607 g, 83%) as a trifluoroacetic acid salt. MS (ESI): M+l = 542.
F.
The amine (0.025 g, 0.038 mmol, 1 eq) from step E was combined with diisopropylethylamine (0.0146 mL, 0.038 mmol, 2.2 eq) in dichloromethane (1.5 mL) under Argon. The solution was treated with methylchloroformate (0.0028 mL, 0.0362 mmol, 0.95 eq) . After stirring for approximately 10 min., the reaction mixture was applied directly to a 20x20 cm (500 uM, silica gel GF) preparative thin layer chromatography plate and eluted with 95:5 dichloromethane : methanol. The product band was removed from the plate and the product was washed from the silica gel with 85:15 dichloromethane : methanol (10 mL) . The solution was evaporated in vacuo, triturated with hexane, evaporated in vacuo, and dried under high vacuum to provide the carbamate as a white solid (0.0164 g, 72 %) . The product was lyophilized from acetonitrile : water (1:1). MS (APCI) : M + Na = 622. H NMR (CDC13 + NaOD) : 1.66 (m, IH) ; 1.90 (m, 3H) ; 2.29 (m, IH) ; 2.64 (m, IH) ; 2.92 ( , 7H) ; 3.18 (m, IH) ; 3.40 (m, IH) ; 3.56 (s, 3H) ; 3.66 (m, IH) ; 3.85 (m, IH) ; 3.98 ( , IH) ; 4.26 (m, IH) ; 5.27 (m, IH) , 6.07 (d, IH, J=7.8); 7.14 (m, 6H) ; 7.28 (m, 8H) .
Example ?6
Synthesis of Compound 236
Figure imgf000276_0001
The aminomethyl pyrolidinone (0.025 g, 0.038 mmol, 1 eq) was combined with diisopropylethylamine (0.0146 mL, 0.038 mmol, 2.2 eq) in dichloromethane (1.5 mL) and cooled to -78 °C with a dry ice acetone bath. The solution was treated with trifluoromethane sulfonic anhydride (0.0064 mL, 0.038 mmol, 1 eq) in dichloromethane (0.5 mL) . The reaction mixture was then allowed to warm to room temperature and applied directly to a 20x20 cm (500 uM, silica gel GF) preparative thin layer chromatography plate and eluted with 95:5 dichloromethane : methanol. The product band was removed from the plate and the product was washed from the silica gel with 85:15 dichloromethane : methanol (10 mL) . The solution was evaporated in va cuo to a residue and lyophilized from acetonitrile : water (1:1) to provide the desired product as a white lyophile (0.008 g, 31 %). MS (APCI): M + Na = 696. H NMR (CDC13 + NaOD) : 1.64 (m, IH) ; 1.98 (m, 3H) ; 2.22 (m, IH) ; 2.72 (m, IH) ; 2.91 (m, 8H) ; 3.47 (m, IH) ; 3.78 (m, IH) ; 3.97 (m, IH) ; 4.09 ( , IH) ; 4.31 (m, IH) ; 5.23 (m, IH) ; 6.17 (d, IH, J=8.7); 7.21 (m, 14H) .
Example $1
Synthesis of Compound 211
Figure imgf000277_0001
The aminomethyl pyrolidinone (0.030 g, 0.046 mmol, 1 eq) was combined with diisopropylethylamine (0.0175 mL, 0.10 mmol, 2.2 eq) and 3- (R)-hydroxy-tetrahydrofuran-N- hydroxysuccinimide carbonate (W093-US8458, 0.016 g, 0.046 mmol, 1 eq) in dichloromethane (1.5 mL) and allowed to stir for 16 h at ambient temperature. The dichloromethane was removed in vacuo and replaced with acetonitrile (2 mL) . The mixture was heated at reflux for 20 min. and then cooled and evaporated in vacuo . The residue was dissolved in dichloromethane (~0.5 mL) , applied directly to a 20x20 cm (500 uM, silica gel GF) preparative thin layer chromatography plate and eluted with 9:1 dichloromethane : methanol. The product band was removed from the plate and the product was washed from the silica gel with 85:15 dichloromethane : methanol (10 mL) . The solution was evaporated in vacuo to a residue and lyophilized from acetonitrile : water (1:1) to provide the desired product as a white lyophile (0.022 g, 73 %) . MS (ESI) : M + Na = 678. H NMR (CDC13 + NaOD) : 1.65 ( , IH) ; 1.93 (m, 5H) ; 2.32 (m, IH) ; 2.65 ( , IH) ; 2.90 (m, 7H) ; 3.22 (m, IH) ; 3.37 (m, IH) ; 3.54 (m, 2H) ; 3.79 (m, 4H) ; 3.97 (m, IH) ; 4.22 (m, IH) ; 5.11 (m, IH) ; 5.27 (m, IH) ; 6.34 (d, IH, J=8.9) ; 7.22 (m, 14H) .
Example 98
Figure imgf000278_0002
Figure imgf000278_0001
2. HCl
The starting cyclic urea was obtained following procedures outlined in Examples 11 and 12. Coupling with the epoxide followed the protocol detailed in Example 24. Example 99
Synthesis of Compound 242
Figure imgf000279_0001
0.3g of the protected intermediate obtained in Example 98 was treated with 10 mL of TFA over 5 h at room temperature. The reaction was quenched by removing the TFA, and the resulting crude treated with excess of sodium carbonate in methanol/water for 10 minutes. The solvents were removed, product extracted between ethyl acetate/water, organics combined, dried with magnesium sulfate, removed in vacuo, and purified by preparative HPLC, resulting in 0.15g (76.7%) of product 2. XH NMR (CDC13, 300 MHz) δ 8.10 (IH, d, J=8.4), 7.24 (10H, m) , 7.05 (5H, m) , 5.28 (m, IH) , 4.10 (IH, t, J=4.2), 3.97 (IH, t, J=4.9), 3.53 (IH, m) , 3.39 (2H, m) , 2.95 (5H, m) , 2.69 (m, 2H) , 2.54 (IH, dd) , 2.17 (IH, m) , 1.92 (IH, m) , 1.78 (m, IH) . Low resolution MS m/e 514.1 (M+H+), m/e 536.2 (M+Na+) .
Example 100
Synthesis of Compound 243
KOtBu R-halide
Figure imgf000280_0001
Figure imgf000280_0002
A solution of 20 mg (0.039 mmol) of the urea obtained Example 99 in 1 mL DMF was treated with potassium t- butoxide (26.3 mg, 0.234 mmol, 6 equiv) and equilibrated at room temperature for 10 min. Next, 6.3 mg of 3-picolyl chloride in 1 mL DMF was added and the reaction quenched after 20 min. Solvent were then removed and the residue purified on preparative RP HPLC resulting in 14.2 mg (60.20%) of the product. 1H NMR (d6-acetone, 400 MHz) δ 8.57 (d, IH, J=5.3), 8.42 (s, IH) , 8.01 (d, IH, J=8.0), 7.80 (t, IH, J=5.9), 7.20 ( , 14H), 6.92 (d, IH, J=8.8), 5.23 ( , IH) , 4.29 (d, IH, J=16.2), 4.29 (m, IH) , 4.11 (d, IH, J=16.2), 3.98 (m, 2H) , 3.48 (dd, IH) , 3.18 (m, 2H) , 3.00 (m, 4H) ,
2.75 (m, 3H) , 1.93 (m, IH) , 1.88 (m, IH) , 1.66 (m, IH) . Low resolution MS m/e 605.4 (M+H+) , m/e 627.4 (M+Na+) .
Example 101
Synthesis of Compound 244
KOtBu R-halide
Figure imgf000281_0001
Figure imgf000281_0002
This compound was synthesized using the protocol outlined for Example 100 starting from 51 mg (0.1 mM) of cyclic urea and 3-methylbenzyl bromide (18.5 mg, 0.1 mmol, 1 equiv), resulting in 6.2 mg of the product after preparative HPLC purification. H NMR (d6-DMSO, 300 MHz) δ_7.68 ( H, d, J=8.5), 7.24 (19H, m) , 5.18 (IH, m) , 4.26 (IH, m) , 4.16 (IH, d, J=15.7), 4.01 (IH, d, J=15.7), 3.79 (2H, m) , 3.33 (IH, m) , 3.05 (6H, m) , 2.78 (m, 2H) , 2.62 (2H, m) , 2.24 (s, 3H) , 1.80 (IH, m) , 1.38 (IH, m) . Low resolution MS m/e 618.2 (M+H+) .
Example 102
Synthesis of Compound 245
KOtBu R-halide
Figure imgf000281_0003
Figure imgf000281_0004
This compound was synthesized using the protocol outlined for Example 100 starting from 51 mg (0.1 mM) of cyclic urea and 3-fluorobenzyl bromide (18.9 mg, 0. mmol, 1 equiv), resulting in 7.6 mg of the product after preparative HPLC purification. H NMR (d6-DMS0, 300 MHz) δ 7.71 (IH, .d, J=8.5), 7.24 (19H, m) , 5.18 (IH, ) , 4.28 (IH, m) , 4.19 (IH, d, J=15.7), 4.03 (IH, d, J=15.7), 3.82 (2H, m) , 3.33 (IH, dd) , 3.05 (6H, m) , 2.80 (m, 2H) , 2.59 (2H, m) , 1.79 (IH, m) , 1.38 (IH, m) Low resolution MS m/e 622.1 (M+H+) .
Example 103
Synthesis of Compound 262
Figure imgf000282_0001
Obtained following the protocol outlined for Example 100 using 2-picolyl chloride. LC/MS-MH+ 605.
Example 1Q4
Synthesis of Compound 213
KOtBu R-halide
Figure imgf000283_0001
Figure imgf000283_0002
Obtained following the protocol outlined for Example 100 using 3, 4, 5-trimethoxybenzyl chloride. λU NMR (DMS0)d6 1.35 (t,lH),1.78 (t,lH), 2.45 (m,2H), 2.62 (m,2H), (s, 6H) , 3.8 (m,2H), 4.1 (q,2H), 4.28 (t,2H), 5.18 (m,2H), 6.45 (s,2H), 6.93-7.38 (m,16H) 7.68 (d,2H), LC/MS-MH+ 694.
Example 105
Synthesis of Compound 246
Figure imgf000283_0003
Obtained following the protocol outlined for Exampl*
100 using 4-amidobenzyl chloride.
1H NMR (DMS0d6) 1.35 (t,lH),1.78 (t,lH) ,2.45
(m,2H) ,2.62 (m, (s,6H), 3.8 (m,2H), 4.1(q,2H), 4.2' (t,2H), 5.18 (m,2H), 6.45 (s,2H), 6.93-7.38 (m,16H) 7.68 (d,2H), LC/MS-MH+ 694.
Example 106
Synthesis of Compound 257
Figure imgf000284_0001
Following the procedure outlined in Example 21 the desired ketoamide was obtained as a white fluffy solid after purification on reversed phase HPLC. M+H: 504 λli NMR: 1.38 and 1.48 (9H,s), 1.8-3.0(ca 7H,m), 3.72 and 3.73 (3H,s), 3.5(lH,m), 3.8(lH,m), 4.0(2H,m), 4.2- 4.8(3H,m) 7.2- .4 (5H,m) . Note: Complex NMR signals due to rotational isomers, diastereomers and ketone-hydrate equillibria.
Example 107
Synthesis of Compound 258
Figure imgf000284_0002
The procedure was followed as described in Example 21 except that instead of thioprolme-t-butylamide being coupled to the ketoacid 1, thioprolme-dimethyl propargylamide 2 was used. This compound was made by treating a 0 °C solution of N-BOC-4-thιo-L-prolιne (Sigma, 2.0 g, 8.6 mmol) in THF (40 mL) with diisopropylethylam e (4.5 mL, 26 mmol) followed by dropwise addition of isobutyl chloroformate (1.1 mL, 8.6 mmol) via syringe. The reaction was stirred for 30 minutes at 0 °C before the dropwise addition of 90*
1, 1-dιmethylpropargylamme (Aldrich, 1.0 mL, 8.6 mmol) . After stirring for 17 h at room temperature, the reaction was concentrated in vacuo. Ethyl acetate (70 mL) and water (35 mL) were added to the residue and the layers were partitioned. The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo. The crude residue was then dissolved dichloromethane (20 mL) and treated slowly with trifluoroacetic acid (20 mL) . The reaction was stirred for 24 h before being diluted with ethyl acetate (70 mL) and carefully neutralized with 10% sodium carbonate to pH 7. The layers were partitioned and the organic layer was dried over sodium sulfate, filtered, and concentrated m vacuo. Flash chromatography over silica gel (1:1 hexane:ethyl acetate) gave amide 2 as a white foam. MS (ES+) = 199 (M+l) . Coupling of ketoacid 1 (300 mg, 0.854 mmol) with amide 2 (170 mg, 0.854 mmol) gave ketoamide 3 (84 mg, 0.211 mmol, 25%) after preparatory silica gel TLC (3:1 ethyl acetate:hexane) . MS (AP+) = 532 (M+l), 554 (M+Na) ;
XHNMR (CDC13) : d 1.66 (s, 3H) , 1.69 (s, 3H) , 1.94 (m, 2H), 2.37 (d, IH) , 2.51 (m, 3H) , 2.92 (m, IH) , 3.21 ( , 2H) , 3.52 (m, IH) , 3.83 (m, IH) , 4.22 (m, 2H) , 4.44 (m, IH) , 4.81 (m, IH) , 5.00 (m, IH) , 6.57 (d, IH) , 7.1 (m, 4H) , 7.22 (m, 6H) .
Example 109
Synthesis of Compound 263
1.) sodium hydride, DMF
Figure imgf000286_0001
Figure imgf000286_0002
Figure imgf000286_0003
rt
Prepared using the procedure outlined in Example 24. The acetonide was purified by column chromatography: 65/35 hexane/ethyl acetate. MS: M+Na = 643. The product was purified by column chromatography: 40/60 hexane/ethyl acetate. MS: M+Na = 603 1H NMR (CDC13) l-05(m, IH) ; 1.10-1.40 (m, 6H) ; 1.50-1.75 (m, 4H) ; 1.80- 2.00(m, 2H) ; 2.45(m, IH) ; 2.80-3.10 ( , 4H) ; 3.20 (m, 2H) ; 3.30 ( , IH) ; 3.45(s, IH) ; 3.65(m, IH) ; 3.80(m, IH) ; 3.90 (m, IH) ; 4.25(m, IH) ; 4.60 (m, IH) ; 5.27 (m, IH) ; 6.00(d, IH) ; 7.10-7.40 (m, 14H) .
Example 109
Synthesis of Compound 206
A.
Figure imgf000287_0001
A solution of 22.3g (0.147 mol, 1 equiv) of S(-)-2- Amino-3-phenyl-l-propanol in 30 L THF, cooled to 0 °C, was treated with 25.5 mL (0.147 mol, 1 equiv) of DIEA, followed by addition of 11.7 L (0.147 mmol, 1 equiv) of chloroacetyl chloride. After 1 hr at room temperature, 18. Og (0.16 mol) of potassium - tert- butoxide was added at 0 °C, the reaction warmed up to room temperature and allowed to proceed for 15 min. Solvents were then removed and the crude residue partitioned between ethyl acetate/water, organics dried over MgS04 resulting in 23.8g (85%) of the desired product. ^Η NMR (CDCL3, 300 MHz) δ 7.20 (m, 5H) , 6.
(s, IH) , 4.15 (m, 2H) , 3.75 (m, IH) , 3.86 (dd, IH,
J 1==1111..66,, 33..77)),, 33..5555 ( [dddd,, IIHH,, JJ==l1l1..(6, 6.3), 2.82 (m, 2H) .+ , Low resolution MS m/e 192.1 (M+H
B. A solution of 0.477g (2.5 mmol, 1 equiv) of the morpholinone above in 1 mL of anhydrous DMF was treated with 12 mg (0.5 mmol, 0.2 equiv) of sodium hydride (95%) at O'C. The reaction was continued at room temperature for 10 min, and then cooled down tc O'C, followed by addition of 0.813g (2.5 mmol, 1 equiv) of epoxide in 1 L DMF. The reaction was then carried out at 50'C for 5 h. Following ethyl acetate/water extraction, the organics were combined and dried resulting in 1.18 g of crude product, used further without purification. Low resolution MS /e 539.0 (M+Na+) .
C.
A solution of 1.18g (2.287 mol, 1 equiv) of the above crude in 4 mL anhydrous THF was treated with 0.44g (3.43 mol, 1.5 equiv) of DIEA, followed by 0.907g (3.43 mmol, 1.5 equiv) of TBDMS triflate. After 1 h at room temperature, the product was purified on silica gel
(Rf=0.26, 1:3 ethyl acetate/hexane), yielding 0.85g of the TBDMS ether (59.0%) . 1H NMR (CDCL3, 300 MHz) δ 7.50 (d, 2H, J=8.9), 7.24 (m, 5H) , 6.97 (d, 2H, J=8.9), 4.40 (m, IH) , 4.21 (d, IH, J=6.3) , 4.17 (d, IH, J=6.3), 3.82 (s, 3H) , 3.65 (m, 2H) , 3.54 (m, IH) , 3.35 ( , IH) , 3.17 (m, IH) , 3.00 (m, 4H) , 2.77 (m, IH) , 2.21 ( , IH) , 1.79 (m, IH) , 1.57 (m, 5H) , 1.24 (m, IH) , 1.03 ( , IH) , 0.86 (s, 9H) , 0.05 (s, 3H) , 0.02 (s, 3H) . Low resolution MS m/e 653.1 (M+Na+) , m/e 631.1 (M+H+) .
D.
A solution of 0.12g (0.19 mmol, 1 equiv) of the precursor above in 1.5 mL THF was cooled to -78 "C and added 0.25 mL (0.25 mmol, 1.3 equiv) lithium bis (trimethylsilyl) amide (IM solution in THF) . After 20 min, 0.029 mL (0.248 mmol, 1.3 equiv) of benzyl bromide was added and reaction allowed to proceed at room temperature for additional 1 h. Purification on silica gel (mixture of diastereomers, Rf=0.46, 0.51 in 1:3 ethyl acetate/hexane) provided 44 mg (32.2%) of the TBDMS-protected product. Low resolution MS m/e 1464.6 (2M+Na+), m/e 721.1 (M+H+).
E.
A solution of 40 mg of the silylated product above in
0.3 mL THF was treated with 0.3 mL of IM TBAF in THF for 25 min at room temperature and purified on a silica column, resulting 30 mg of the final product. Rf=0.38 and 0.34 (2/5/0.3 ethyl acetate: hexane: methanol) . XH NMR (CDCL3, 300 MHz) shows both diastereomers and integrates as expected. Low resolution MS m/e 629.3 (M+Na+) .
Example 110
Synthesis of Compound 205
Figure imgf000289_0001
A solution of 0.12g (0.19 mmol, 1 equiv) of the compound prepared m Example 109C was dissolved 1.5 mL THF and was treated with 0.30 mL (0.30 mmol, 1.5 equiv) of lithium bis (trimethylsilyl) amide (IM solution in THF) at -78 °C. After 20 min, 0.023 mL (0.266 mmol, 1.4 equiv) of allyl bromide was added, reaction allowed to warm up to the room temperature and carried out for additional 1 h. The reaction was then quenched with aqueous ammonium chloride and both diastereomers separated on a silica gel. The (lower) Rf=0.50 diastereomer (1:3 ethyl acetate/hexane) was then treated with 10-fold excess of TBAF (IM m THF) for 25 mm at room temperature, followed by another silica purification, which provided 14 mg of the desired allylated product. 1H NMR (CDCL3, 300 MHz) δ 7.73 (d, 2H, J=9.0), 7.24 (m, 5H) , 6.99, (d, IH, J=8.9), 5.84 ( , IH) , 5.12 ( , 2H) , 4.27 (m, IH) , 4.10 (m, IH) , 3.86 (s, 3H) , 3.84 ( , 3H) , 3.58 (m, IH) , 2.8-3.3 (m, 7H) , 2.62 (m, 2H) , 2.09 (m, IH) , 1.60 (m, 6H) , 1.24 ( , 2H) . Low resolution MS m/e 579.3 (M+Na+), m/e 1135.4 (2M+Na+) .
Example 111
Synthes i s o f Compound 207
Figure imgf000291_0001
A solution of 0.092g of the morpholinone described in Example 20 (0.35 mmol, lequiv) in 1.5 L anhydrous DMF was cooled to 0 °C and added 9.6 mg (0.4 mmol, 1 equiv) of NaH. After l/2h 0.13g (0.32 mmol) of the epoxide 2 was added and reaction carried out at room temperature for 10 h, quenched with IN HClaq, and purified on preparative RP HPLC. Yield 70 mg (36.5%) . Low resolution MS m/e 622.1 (M+Na+), m/e 1221.1 (2M+Na+)
Exa ple 2
Using the methods described by Pennmgton et al . and Partaledis et al . (supra), we obtained inhibition constants for the following compounds of this invention:
Compound κ_ (nM)
1 160
2* 180 3* 1,800
5* >10,000 6* >10 , 000
7 9
8* 5
9* 90
5 10 >10 , 000
11 >10,000
12 >10,000
13 225
14 16 10 15 550
16 56
17 115
18 15
19 3,000 15 20 1.5
21 >20,000
22 600
23 70
24 350 20 25 83
26 58
27 3,000
28 1,400 30 >15,000
25 31 390
32 160
33 1,100
34 950
35 130 30 36 >20,000
37 >20,000
38 17 39
40
41
42
43
44
45
Figure imgf000293_0001
47
10 50*
51*
52*
54*
55*
15 56*
57*
58*
59*
60*
20 61*
62*
63*
64*
65*
25 66*
67*
68*
69*
70*
30 71*
72*
73*
Figure imgf000293_0002
74* 1,200
76* 250
77* 560
78* 10
5 79* >3,000
80* 3
82* 15
83* 0.50
85* 2,600
10 87* 15
88* 270
90* 220
91* 12
92* (isomer 1) 3.0
15 92* (isomer 2) 300
93* 420
95* 10
96* 4
98* >10,000
20 102* 1,200
105* >10,000
109* 250
111* >10,000
112* 8,600
25 113* >10,000
114* >1,000
115* >10,000
123* (isomer 1) 300
123* (isomer 2) 13
30 124* (isomer 1) 800
124* (isomer 2) 1900
125* (isomer 1) 400 10
15
20
25
30
Figure imgf000295_0001
228* 16
229* 28
232* 500
233* (isomer 1) 23 233* (isomer 2) 1200
235* 270
236* 3.6
* Inhibition constant measureα at pH 6.0.
Example 113
Using the MT4 cell assay method (supra), we measured the antiviral activity for the following compounds of this invention:
J 50 I-M)
16
9
1.0 0.32 0.21 2
0.40 0.90 0.74 0.85 1.0
2.4 2.9
Figure imgf000296_0001
0.75 2.15
0.4
1.7
6.0
0.10
0.68
2.0
2.0
3.5 2.75
0.48 0.79 2.47 3.7 1.6
Figure imgf000297_0001
5.0
The above data show that each of the tested compounds inhibits HIV aspartyl protease. While we have described a number of embodiments of this invention, it is apparent that our basic constructions may be altered to provide other embodiments which utilize the products and processes of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims, rather than by the specific embodiments which have been presented by way of example.

Claims

We Claim:
A compound according to formula I
Figure imgf000298_0003
Figure imgf000298_0001
wherein : each Z is
Figure imgf000298_0002
wherein any Z is optionally fused with R ; each X and X' is independently selected from the group consisting of -C(0)-, -C(0)C(0)-, -S(0)- and -S (0) 2; each Y and Y' is independently selected from the group consisting of -(C(R 2) )p _, -NR2 p" -, -(C(R2)2)p-M-, >C=C(R2)2, and -N(R2)-CH2-; each R is independently selected from the group consisting of hydrogen; R ; Cι_-Cg alkyl; C2-Cg alkenyl; C -Cg alkynyl; C3-C6 cycloalkyl optionally fused with R6; C5-C6 cycloalkenyl optionally fused with
R 6; and when two R1's are attached to adjacent atoms, the two R1 ' s together with their attached adjacent atoms form a carbocyclic or heterocyclic ring system which is optionally fused with R ; wherein any member l 2 of R is optionally substituted by one or more R ;
2 each R is independently selected from
3 hydrogen; R ; Ci-Cg alkyl; C2-Cg alkenyl; C2-C6 alkynyl;
C3~C6 cycloalkyl optionally fused with R ; C5-C6 g cycloalkenyl optionally fused with R ; and when two
R 2 's are attached to the same gemmal atom, the R2 's together with their attached gemmal atom form a spirocarbocyclic or spiroheterocyclic ring system;
2 wherein any member of R is optionally substituted by one or more R ;
3 each R is independently selected from oxo, OR9, N(R9)2, N(R9)-X-R9, N(R9)-X-0R9,
N(R9)-X-N(R9)2, SR9, X-R9, 0-X-N(R9)2, C(0)N(R9)2,
9 6 halogen, N02, CN, COOR and R ;
4 each R is independently selected from from the group consisting of OR 9; N(R9)2; X-R9;
C(0)N(R9)2; R6; C_-C _ alkyl; 2-C^ alkenyl; C3-C6 cycloalkyl optionally fused with R ; C5~C6 cycloalkenyl
6 4 optionally fused with R ; wherein any member of R is optionally substituted by one or more groups independently selected from R 9 or R3; each R is independently selected from the group consisting of H, OH, 0 and R ; each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected from the group consisting of oxo, -OR9, -R9, -N(R9) (R9),
9 9 9 9 9 9 9
-N(R )-X-R , SR , -X-R , -0-X-N(R )2, -R -OR , -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3;
7 each R is independently selected from the group consisting of hydrogen, OH and O; p each R .is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, and heterocyclyl;
9 each R is independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heterocyclyl, aralkyl, carbocyclylalkyl and heterocyclylalkyl; wherein any aryl, carbocyclyl or heterocyclyl is optionally fused
8 8 with R and wherein any member of R is optionally substituted by one or more groups independently selected from -OR8, -N(R8)2, -CN, -N02, -X-R8, -X- N(R8)2 -C(0)OR8, -N(R8)-XN(R8)2, or halogen; each Q is independently selected from the group consisting of CH and N; each M is independently selected from the
2 group connssiisstting of NH, -NR -, -0-, -S-, -S(O)- and -S(0)2-; each n is independently 1 or 2; each r is independently 0, 1 or 2; each p is independently 1 or 2; each q is independently 1, 2 or 3; and each G is independently selected from the
2 group consisting of -NH-, -NR -, -0-, -S-, -S(O)-,
S(0)2, -C(O)-, and -C(R2)2-.
2. The compound according to claim 1, wherein: each Y and Y' is independently selected
2 2 from the group consisting of -(C(R ) )p~, -NR -,
- (C(R2)2)p-M-, and -N(R2)-CH2-; and
3 each R is independently selected from oxo, OR9, N(R9)2, N(R9)-X-R9, N(R9)-X-OR9, SR9, X-R9,
0-X-N(R9)2, C(0)N(R9)2, halogen, N02, CN, COOR9 and R6
3. The compound according to claim 1 having the structure of formula IA:
Figure imgf000301_0001
wherein : each R 12 is independently selected from the group consisting of R ; C^-Cg alkyl optionally substituted with R ; C -C6 alkenyl; C -Cg alkynyl; C3-Cg cycloalkyl optionally fused with R ; C5-Cg cycloalkenyl optionally fused with R 6; wherein any member of R12 is
2 optionally substituted by one or more R .
4. The compound according to claim 1, wherein n is 1.
5. The compound according to claim 1 having the structure of formula II:
Figure imgf000302_0001
6. The compound according to claim 1 having the structure of formula III:
Figure imgf000302_0002
(III)
7. The compound according to claim 1, wherein:
X is -C(O)- or -S(0)2-" and
Y is -(C(RP2)p-M-.
The compound according to claim 1 , wherein:
X is -C(O)- or -S(0)2-; and Y is (-C(R2)2-)p. 9. The compound accordmg to claim 1, wherein:
X is -C(O)-, -C(0)C(0)- or -S(0)2-; and Y is -N(R2)- or -N(R2)-CH2-.
10. A compound according to formula IV:
Figure imgf000303_0001
wherein:
X and X' are independently -C(O)- or
-S(0) -;
Y is -(C(R2)2)-M-, -(C(R2)2)p-/ -N(R2)- or
-N(R2)-CH2-; and each R 1, R2, R7, R4, p and M is independently as defined in claim 1.
11. A compound accordmg to formula V:
Figure imgf000304_0001
wherein:
X is -C(O)- or -S(0)2-;
Y is -(C(R2)2)-M-, -(C(R2)2)p-, -N(R2)- or
-N(R2)-CH2-;
R10 is 0 or H2; each R is independently H, OH or 0, wherein both R are not simultaneously hydrogen; Z is a structure of formula VI:
Figure imgf000304_0002
(VI)
wherein any structure of formula VI is optionally fused with an aryl, carbocyclic or heterocyclic ring and is optionally substituted with 1-3 substituents
2 independently selected from R ; and each R1, R2, R , R4, R8, p, q, G, M, Q and X' is independently as defined in claim 1. 12. The compound according to claim 11, wherein „R10 and. „R11 are 0.
13. The compound according to claim 12, wherein: q is 1;
G is S; and
X' is -C(0)-.
14. The compound according to claim 13,
4 wherein R is t-butylamino.
15. The compound according to claim 12, wherein:
X is -C(O)-;
Y is -(C(R ) 2P'pn-- and
R7 is H.
16. The compound according to claim 11, wnerein:
X and X' is -C(0)-; Y is -(C(R2)2)-; R7 is H;
R is H2; and
11 11 one R is H and one R iε OH.
17. The compound according to claim 16, wherein R within the definition of Y is selected from
3 hydrogen, R or C^-Cg alkyl optionally substituted with
R3. 18. The compound according to claim 17,
2 wherein R within the definition of Y is selected from g hydrogen, -N(R )2 or heterocyclyl, which may be optionally benzofused, and wherein said heterocyclyl may be optionally substituted with one or more groups selected from the group consisting of oxo, -OR 9, -R9,
9 9 9 9 9 9 9 9
-N(R ) (R ) , -N(R )-X-R , SR , -X-R , -0-X-N(R )2, -R -
OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3.
19. The compound according to claim 18, wherein at least one R 2 -within the definition of Y is selected from the group consisting of:
Figure imgf000306_0001
Figure imgf000306_0002
Figure imgf000306_0003
\=J
Figure imgf000307_0001
Figure imgf000307_0002
Figure imgf000308_0001
Figure imgf000308_0004
Figure imgf000308_0003
20. The compound according to claim 17,
2 wherein at least one R within the definition of Y is aryl optionally substituted with one or more groups selected from the group consisting of oxo, -OR 9, -R9,
-N(R9) (R9), -N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-
OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3
21. The compound according to claim 17, wherein at least one R within the defini .tt:ion of Y is
Cι_-Cg alkyl optionally substituted with R,3~ 22. The compound according to claim 21, 3 wherein at least one R within the definition of Y is pyridyl, triazolyl, oxazolyl, isoxazolyl, pyrimidyl, pyrazolyl, pyridazinyl, thiazolyl, imidazolyl, thienyl thiadiazolyl, oxadiazolyl, triazinyl or pyrazinyl
3 wherein said R may be optionally substituted with 1-3 substituents selected from -OR9, -R9, -N(R9) (R9), -
9 9 9 -9 9 9 9
N(R )-X-R , SR , -X-R , -0-X-N(R )2, -R -OR , -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3.
23. The compound according to claim 21, 3 wherein R within the definition of Y is aryl optionally substituted with 1-3 substituents selected from -OR9, -R9, -N(R9) (R9), -N(R9)-X-R9, SR9, -X-R9,
-0-X-N(R9)2, -R9-OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3.
24. The compound according to any one of claims 17-23, wherein R is benzyl; and Z is
Figure imgf000309_0001
25. The compound according to any one of claims 17-23, wherein R is benzyl optionally substituted with 1-3 substituents selected from -OR9, -N(R9) (R9), SR9, -X-R9, -R9-0R9, -CN, halogen, -N02, and
-CF3.
26. The compound according to claim 25, wherein Z is
Figure imgf000310_0001
27. The compound according to claim 25, wherein R is benzyl optionally substituted with 1-3 substituents selected from the group consisting of 0CH3, OH and NH2.
28. The compound according to claim 27, wnerem Z is
Figure imgf000310_0002
29. A compound accordmg to formula V, wherein:
Figure imgf000311_0001
each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with one or more groups selected
9 9 9 9 from the group consisting of oxo, -OR , -R , -N(R ) (R ),
-N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, -CF3, -O- (CH2) q-R6,
9 -0-(CH2)q-OR , 2, 3-methylenedιoxy and 3,4- methylenedioxy; and
1 2 3 4 5 7 8 9 each X, X', Y, Y', Z, R , R , R , R , R , R , R , R , Q,
M, n, r, p, q and G is independently as defined in claim 1.
30. The compound accordmg to claim 29,
2 wherein R with the definition of Y is selected from
3 hydrogen, R or Cι_-Cg alkyl optionally substituted with
R3.
31. The compound accordmg to claim 11, wherein:
Figure imgf000311_0002
Y is -N(R2)-;
R7 is H;
R is H2; and one R is H and one R is OH. 32. The compound according to claim 11, wherein:
X and X' is -C(O)-;
Y is - (C(R2)2)-M-;
M is 0;
R7 is H; .
R is H ; and one R is H and one R is OH.
33. The compound according to claim 1, having the structure of formula IX:
Figure imgf000312_0001
(IX)
wherein:
X is -C(O)- or -S(0)2-
34. The compound according to claim 33, wherein:
X is -C(O)-;
Y is - (C(R2)2)-M-; and
R7 is H. 35. The compound accordmg to claim 33, wherein:
X is -C(O)-;
Y is -N(R2)-; and
Figure imgf000313_0001
36. The compound accordmg to claim 33, wherein:
X is -C(O)-;
Y is -(C(R2) P-; and
Figure imgf000313_0002
37. The compound accordmg to claim 1, having the structure of formula XII:
Figure imgf000313_0003
(XII)
wherein:
X and X' are independently -C(O)- or
-S(0)2-.
38 . The compound accordmg to claim 37 , 4 wherein R is l-ammo-2-hydroxymdanyl . 39. The compound accordmg to claim 1, having the structure of formula XIII:
Figure imgf000314_0001
wherein:
X and X' are independently -C(O)- or
-S(0)2-
40. The compound according to claim 39, wherein:
X' is -C(0)-
Y is - (C(R or -N(R )-; and
R is H.
41. The compound according to claim 40, wherein:
X is -C(O)-; and Y is - (C(R2) ,)-.
42. The compound according to claim 41,
2 wherein R within the definition of Y is selected from
3 hydrogen, R , or C-j_-C6 alkyl optionally substituted with
R3. 43. The compound according to claim 42,
2 wherein R within the definition of Y is selected from
9 hydrogen, -N(R )2 or heterocyclyl, which may be optionally benzofused, and wherein said heterocyclyl may be optionally substituted with 1-3 groups selected
9 9 9 9 from the group consisting of oxo, -OR , -R , -N(R ) (R ),
-N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, and -CF3.
44. The compound according to claim 43,
2 2 wherein at le»aasstt oonnee RR wwiitthhiinn tthhee ddeeffinition of Y is selected from the group consisting of:
Figure imgf000315_0001
Figure imgf000316_0001
Figure imgf000316_0002
Figure imgf000316_0005
Figure imgf000316_0003
*V
Figure imgf000316_0004
Figure imgf000317_0001
Figure imgf000317_0002
45. The compound according to claim 42, 2 wherein at least one R within the definition of Y is aryl optionally substituted with one or more groups selected from the group consisting of oxo, -OR9, -R9,
9 9 9 9 9 9 9 9
-N(R ) (R ), -N(R )-X-R , SR , -X-R , -0-X-N(R )2, -R -
OR9, -CN, -C02R9, -X-N(R9) (R9) , halogen, -N02, ana -CF3.
46. The compound accordmg to claim 42,
2 wherein at least one R withm the definition of Y is
3 Cτ_-Cg alkyl optionally substituted with R .
47. The compound accordmg to claim 46, 3 wherein at least one R within the definition of Y is pyridyl, triazolyl, oxazolyl, isoxazolyl, pyrimidyi, pyrazolyl, pyridazmyl, thiazolyl, imidazolyl, thienyl thiadiazolyl, oxadiazolyl, triazmyl or pyrazinyl
3 wherein said R may be optionally substituted with 1-3
9 9 9 9 substituents selected from -OR , -R , -N(R ) (R ), -
9 9 9 9 9 9 9
N(R )-X-R , SR , -X-R , -0-X-N(R )2, -R -OR , -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, or -CF3.
48. The compound according to claim 46, wherein R withm the definition of Y is aryl optionally substituted with 1-3 substituents selected from -OR9, -R9, -N(R9) (R9), -N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN, -C02R9, -X-N (R9) (R9) , halogen, -N02, or -CF3
49. The compound accordmg to any one of claims 42-48, wherein: each R is benzyl; and
9 each R not withm the definition of Y is
2-hydroxy danyl 50. The compound accordmg to any one of claims 42-48, wherein each R is independently selected from benzyl optionally substituted with 1-3
9 substituents selected from -OR , -N(R ) (R" SR" -X-R"
-R9-OR9, -CN, halogen, -N02, and -CF3.
51. The compound according to claim 50,
9 < wherein each RR" not withm the definition of Y is 2 hydroxymdanyl
52. The compound accordmg to claim 50, wherein each RI is independently selected from benzyl optionally substituted with 1-3 substituents selected from the group consisting of OCH3, OH and NH2.
53. The compound according to claim 52,
( wherein each RR" not withm the definition of Y is 2- hydroxymdanyl
54. A compound according to formula XIII, wherein:
Figure imgf000319_0001
each R is independently selected from the group consisting of aryl, carbocyclyl and heterocyclyl, wherein said aryl, carbocyclyl or heterocyclyl is optionally substituted with, one or more groups selected
9 9 from the group consisting of oxo, -OR , -R , -N(R ) (R ),
-N(R9)-X-R9, SR9, -X-R9, -0-X-N(R9)2, -R9-OR9, -CN,
-C02R9, -X-N(R9) (R9) , halogen, -N02, -CF3, -O- (CH2) q-R6,
-0-{CH2)q-0R , 2, 3-methylenedioxy and 3,4- methylenedioxy; and each X, X', Y, Y\ Z, R1, R2, R3, R4, R5, R7, R8, R9, Q,
M, n, r, p, q and G is independently as defined in claim 1.
55. The compound according to claim 54,
2 wherein R within the definition of Y is selected from
3 hydrogen, R or C^-Cg alkyl optionally substituted with
R3.
56. The compound according to claim 40, wherein:
X is -C(O)-; and
Y is -N(R )-.
57. The compound according to claim 40, wherein:
X is -S02-; and Y is -(C(R2)2)-.
58. The compound according to claim 40, wherein
X is -S02-; and Y is -N(R2)-. 59. The compound accordmg to claim 11, wherein:
R is H2; and one R is H and one R is OH; and
Z is selected from the group consisting of:
Figure imgf000321_0001
and R is as defined in claim 1
60. The compound accordmg to claim 11, wherein Z is selected from the group consisting of
Figure imgf000321_0002
Figure imgf000321_0003
one R is H and one R is OH, 61. The compound according to any one of claims 16-32, wherein Z is selected from the group consisting of:
Figure imgf000322_0001
and R is as defined in claim 1.
62. The compound according to any one of claims 16-32, wherein Z is selected from the group consisting of:
Figure imgf000322_0002
63. A compound according to formula I, wherein:
Figure imgf000323_0001
Z is selected from the group consisting of -X'R4, -N(R X'-R , -N(R1)-N(R1)-X'-R4, and formula VI;
Figure imgf000323_0002
(VI)
wherein any structure of formula VI is optionally fused with an aryl, carbocyclic or heterocyclic ring and is optionally substituted with 1-3 members independently
2 selected from R ; and each X, X', Y, Y" R1, R2, R3, R4, R5, R6, R7, R8, R9, Q,
M, n, r, p, q and G is independently as defined in claim 1.
64. The compound of claim 1, selected from the group consisting of compound numbers: 1, 2, 3,
4, 1 , 8, 9, 13, 14, 16, 17, 18, 20, 23, 24, 25, 26, 32,
35, 38, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 62,
63, 72, 75, 76, 78, 80, 82, 83, 91, 92, 94, 95, 96,
101, 102, 109, 121, 122, 123, 124, 126, 127, 128, 129,
131, 132, 133, 134, 135, 137, 138, 140, 141, 145, 146, 147, 149, 150, 155, 156, 160, 161, 162, 164, 165, 170, 171, 175, 176, 177, 179, 180, 185, 186, 190, 191, 192, 194, 195, 200, 201, 208, 219, 220, 228 and 264.
65. The compound of claim 64, selected from the group consisting of compound numbers: 2, 7, 8, 9, 14, 18, 20, 25, 26, 32, 38, 45, 47, 48, 49, 50, 51, 53, 54, 62, 63, 72, 82, 83, 91, 92, 94, 95, 96, 123, 126, 140, 141, 219, 220, 228 and 264.
66. The compound of claim 65, selected from the group consisting of compound numbers: 7, 8, 9, 20, 45, 50, 51, 53, 54, 82, 83, 92, 94, 96, 219, 220, 228 and 264.
67. A pharmaceutical composition comprising an amount of a compound according to claim 1 effective in inhibiting aspartyl protease and a pharmaceutically acceptable carrier, adjuvant or vehicle .
68. The pharmaceutical composition accordmg to claim 67, wherein said pharmaceutical composition is orally administrable.
69. The pharmaceutical composition accordmg to claim 67, further comprising one or more additional agents selected from the group consisting of other anti-viral agents and immunostimulators .
70. The pharmaceutical composition accordmg to claim 69, wherein said other anti-viral agent is a protease inhibitor or a reverse transcriptase inhibitor.
71. The pharmaceutical composition accordmg to claim 70, wherein said protease inhibitor is a HIV protease inhibitor.
72. The pharmaceutical composition according to claim 71, wherein said HIV protease inhibitor or inhibitors are selected from the group consisting of VX-478, saquinavir, mdmavir, ritonavir, nelfmavir, palinavir, U-103017, XM 412, XM 450, BMS 186318, CPG 53,437, CPG 61,755, CPG 70,726, ABT 378 , GS 3333, GS 3403, GS 4023, GS 4035, GS 4145, GS 4234 , and GS 4263.
73. The pharmaceutical composition accordmg to claim 70, wherein said reverse transcriptase inhibitor is a nucleoside analog.
74. The pharmaceutical composition according to claim 73, wherein said nucleoside analog is selected from the group consisting of zidovud e (AZT), dideoxycytid e (ddC) , didanos e (ddl), stavudme (d4T) , 3TC, 935U83, 1592U89 and 524W91.
75. The pharmaceutical composition accordmg to claim 70, wherein said reverse transcriptase inhibitor is a non-nucleoside analog.
76. The pharmaceutical composition according to claim 75, wherein said non-nucleoside reverse transcriptase inhibitor is delavirdme (U90) or nevirapine.
77. The pharmaceutical composition according to claim 67, wherein said pharmaceutical composition further comprises an agent capable of inhibiting the metabolic effects of one or more cytochrome P450 enzyme subtypes.
78. A method for inhibiting aspartyl protease activity comprising the step of contacting an aspartyl protease with the compound accordmg to claim
1.
79. A method for reversibly binding an aspartyl protease comprising the step of contacting the aspartyl protease with the compound according to claim 1, said compound being covalently bound to a solid matrix.
80. A method for preventing HIV infection a mammal comprising the step of administering to said mammal a pharmaceutical composition according to either claim 67 or 68.
81. A method for preventing HIV infection in a mammal comprising the step of administering to said mammal a pharmaceutical composition according to claim 69.
82. A method for treating HIV infection in a mammal comprising the step of administering to said mammal a pharmaceutically effective amount of a pharmaceutical composition according to either claim 67 or 68.
83. A method for treating HIV infection in a mammal comprising the step of administering to said mammal a pharmaceutical composition according to claim 69.
84. The method according to either claim 80 or 82, further comprising the step of administering, to the mammal one or more additional agents selected from the group consisting of other anti-viral agents and immunostimulators via a single or multiple dose.
85. The method according to claim 84, wherein said other anti-viral agent is a protease inhibitor or reverse transcriptase inhibitor.
86. The method according to claim 85, wherein said protease inhibitor is an HIV protease inhibitor.
87. The method accordmg to claim 86, wherein said HIV protease inhibitor is selected from the group consisting of VX-478, saquinavir, indinavir , ritonavir, nelfmavir, palinavir, U-103017, XM 412 , XM 450, BMS 186318, CPG 53,437, CPG 61,755 , CPG 70,726, ABT 378, GS 3333, GS 3403, GS 4023, GS 4035, GS 4145 , GS 4234, and GS 4263. 88. The method according to claim 85, wherein said reverse transcriptase inhibitor is a nucleoside analog.
89. The method according to claim 88, wherein said nucleoside analog is selected from the group consisting of zidovudine (AZT) , dideoxycytidme (ddC) , didanosine (ddl), stavudine (d4T) , 3TC, 935U83, 1592U89 and 524W91.
90. The method according to claim 85, wherein said reverse transcriptase inhibitor is a non- nucleoside analog.
91. The method according to claim 90, wherein said non-nucleoside reverse transcriptase inhibitor is delavirdine (U90) or nevirapme.
92. A method for treating or preventing of viral infection comprising the step of administering to said mammal a pharmaceutical composition accordmg to either claim 67 or 68.
93. A method for treating or preventing HIV related disease effects, including tumors, CMV retinitis, Candida infections, maternal fetal transmission, or AIDS related dementia, comprising the step of administering to said mammal a pharmaceutical composition according to either claim 67 or 68. 94. The composition accordmg to claim 69, wherein the additional anti-viral agents are 3TC and zidovudine (AZT) .
95. The composition accordmg to claim 69, wherein the additional anti-viral agent is 1592U89.
96. A process for preparing a compound of formula XIV:
Figure imgf000329_0001
XIV
wherein R
Figure imgf000329_0002
are defined as in claim 1, comprising the steps of:
(1) reacting a compound of formula XV:
Figure imgf000329_0003
XV
wherein R is defined as in claim 1, in an inert solvent with a base;
(2) reacting the product of step (1) with an aldehyde of R CHO followed by an optional treatment with a dehyratmg agent, wherein R is defined as m claim 1 to give a compound of formula XVI:
Figure imgf000330_0001
wherein R and R are defined as in claim 1;
(3) reacting the product of step (2) in an inert solvent with hydrogen gas in the presence of an hydrogenation catalyst followed by treatment with an anhydrous acid to give a product of formula XIV.
97. A process for preparing a compound of formula XVII:
Figure imgf000330_0002
XVII
wherein R 1 and R2 are defined as in claim 1, comprising the steps of:
(1) reacting a compound of formula XVIII
Figure imgf000331_0001
1 2 wherein R and R are as defined in claim 1, in an inert solvent with a base, then bromomethylacrylic acid;
(2) reacting the product of step (1) with an oxidizing agent;
(3) reacting the product of step (2) in an inert solvent with thioproline t-butylamide and suitable amide-bond coupling reagents to give a product of formula XVII.
98. A process for preparing a compound of formula XIX:
Figure imgf000331_0002
wherein R and r are defined as in claim 1, comprising the steps of:
(1) reacting a compound of formula XX
Figure imgf000332_0001
XX
in an inert solvent with a base, then a bis-leaving group alkane of formula XXI:
Figure imgf000332_0002
wherein LG is selected from halo, arylsulfonate esters and alkylsulfonate esters, and r is defined as in claim 1, to give a product of formula XXII:
Figure imgf000332_0003
wherein R and PG are defined as in formula XX and LG and r are defined as in formula XXI;
(2) reacting the product of step (1) in an inert solvent with a base, to give a product of formula XXIII:
Figure imgf000333_0001
wherein R is defined as in claim 1 and PG is a N- protecting group;
(3) reacting the product of step (2) in an inert solvent with a reagent suitable for removal of the N-protecting group PG to give a compound of formula XIX.
PCT/US1997/001610 1996-01-26 1997-01-22 Aspartyl protease inhibitors WO1997027180A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP97904911A EP0882022A1 (en) 1996-01-26 1997-01-22 Aspartyl protease inhibitors
EA199800661A EA199800661A1 (en) 1996-01-26 1997-01-22 Aspartic protease inhibitors
AU17580/97A AU709239B2 (en) 1996-01-26 1997-01-22 Aspartyl protease inhibitors
JP9527124A JP2000501111A (en) 1996-01-26 1997-01-22 Aspartyl protease inhibitor
BR9707086A BR9707086A (en) 1996-01-26 1997-01-22 Asparta proteinase inhibitors
IL12547997A IL125479A0 (en) 1996-01-26 1997-01-22 Aspartyl protease inhibitors pharmaceutical compositions containing the same and a process for their production
NO983435A NO983435L (en) 1996-01-26 1998-07-24 Disclosed are

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08/592,777 1996-01-26
US08/592,777 US5883252A (en) 1996-01-26 1996-01-26 Aspartyl protease inhibitors
US08/724,563 1996-09-30
US08/724,563 US5945413A (en) 1996-01-26 1996-09-30 Aspartyl protease inhibitors

Publications (1)

Publication Number Publication Date
WO1997027180A1 true WO1997027180A1 (en) 1997-07-31

Family

ID=27081545

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/001610 WO1997027180A1 (en) 1996-01-26 1997-01-22 Aspartyl protease inhibitors

Country Status (12)

Country Link
EP (1) EP0882022A1 (en)
JP (1) JP2000501111A (en)
AP (1) AP844A (en)
AU (1) AU709239B2 (en)
BR (1) BR9707086A (en)
CA (1) CA2243121A1 (en)
CZ (1) CZ234498A3 (en)
HU (1) HUP9901024A2 (en)
IL (1) IL125479A0 (en)
NO (1) NO983435L (en)
PL (1) PL328223A1 (en)
WO (1) WO1997027180A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998052571A1 (en) * 1997-05-17 1998-11-26 Glaxo Group Limited Antiviral combinations containing the carbocyclic nucleoside 1592u89
WO1999033795A1 (en) * 1997-12-24 1999-07-08 Vertex Pharmaceuticals Incorporated Prodrugs of aspartyl protease inhibitors
WO2000009106A2 (en) * 1998-08-14 2000-02-24 Gpi Nil Holdings, Inc. Carboxylic acids and isosteres of heterocyclic ring compounds having multiple heteroatoms for vision and memory disorders
EP1011668A1 (en) * 1997-08-22 2000-06-28 The Wichita State University 1,2,5-thiadiazolidin-3-one 1,1-dioxide derivatives
US6303609B1 (en) * 1998-11-18 2001-10-16 Dupont Pharmaceuticals Company Isoxazoline fibrinogen receptor antagonists
WO2004105750A1 (en) * 2003-05-30 2004-12-09 Neuromed Technologies, Inc. 3-aminomethyl-pyrrolidines as n-type calcium channel blockers
WO2006114001A1 (en) * 2005-04-27 2006-11-02 Ambrilia Biopharma Inc. Method for improving pharmacokinetics of protease inhibitors and protease inhibitor precursors
US7388008B2 (en) 2004-08-02 2008-06-17 Ambrilia Biopharma Inc. Lysine based compounds
JP2008543841A (en) * 2005-06-14 2008-12-04 シェーリング コーポレイション Macrocyclic heterocyclic aspartyl protease inhibitors
US7504517B2 (en) 2006-06-20 2009-03-17 Wyeth Kv1.5 potassium channel inhibitors
US8227450B2 (en) 2005-11-30 2012-07-24 Ambrilia Biopharma Inc. Lysine-based prodrugs of aspartyl protease inhibitors and processes for their preparation
US8410300B2 (en) 2006-09-21 2013-04-02 Taimed Biologics, Inc. Protease inhibitors
CN103626713A (en) * 2012-08-29 2014-03-12 中国科学院上海药物研究所 Aza (oxa)-cyclopentane-2- ketone (thione) compounds, medicine compositions thereof, preparation method and applications
CN105111198A (en) * 2015-08-05 2015-12-02 贵州大学 Nitryl isoxazole spliced 2-pyrrolidone compounds, and preparation method and application thereof
US9701727B2 (en) 2011-06-29 2017-07-11 The Trustees Of Columbia University In The City Of New York Inhibitor of neuronal connectivity linked to schizophrenia susceptibility and cognitive dysfunction

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10047110A1 (en) * 2000-09-22 2002-04-18 Bayer Ag Optically active 2,5-bisaryl-DELTA · 1 · -pyrrolines
EP1699455B1 (en) * 2003-12-15 2013-05-08 Merck Sharp & Dohme Corp. Heterocyclic aspartyl protease inhibitors
JP2005232103A (en) * 2004-02-20 2005-09-02 Nagase & Co Ltd Optically active vicinaldiamine and method for producing the same
GB0412019D0 (en) * 2004-05-28 2004-06-30 Novartis Ag Organic compounds

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994019329A1 (en) * 1993-02-26 1994-09-01 The Du Pont Merck Pharmaceutical Company Substituted cyclic carbonyls and derivatives thereof useful as retroviral protease inhibitors
WO1995024385A1 (en) * 1994-03-07 1995-09-14 Vertex Pharmaceuticals Incorporated Sulphonamide derivatives as aspartyl protease inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994019329A1 (en) * 1993-02-26 1994-09-01 The Du Pont Merck Pharmaceutical Company Substituted cyclic carbonyls and derivatives thereof useful as retroviral protease inhibitors
WO1995024385A1 (en) * 1994-03-07 1995-09-14 Vertex Pharmaceuticals Incorporated Sulphonamide derivatives as aspartyl protease inhibitors

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998052571A1 (en) * 1997-05-17 1998-11-26 Glaxo Group Limited Antiviral combinations containing the carbocyclic nucleoside 1592u89
US6420401B1 (en) 1997-08-22 2002-07-16 Wichita State University 1,2,5, thiadiazolidin-3-one 1,1-dioxide derivatives
EP1011668A1 (en) * 1997-08-22 2000-06-28 The Wichita State University 1,2,5-thiadiazolidin-3-one 1,1-dioxide derivatives
EP1011668A4 (en) * 1997-08-22 2001-08-16 Univ Wichita State 1,2,5-thiadiazolidin-3-one 1,1-dioxide derivatives
WO1999033795A1 (en) * 1997-12-24 1999-07-08 Vertex Pharmaceuticals Incorporated Prodrugs of aspartyl protease inhibitors
WO2000009106A2 (en) * 1998-08-14 2000-02-24 Gpi Nil Holdings, Inc. Carboxylic acids and isosteres of heterocyclic ring compounds having multiple heteroatoms for vision and memory disorders
WO2000009106A3 (en) * 1998-08-14 2000-10-12 Guilford Pharm Inc Carboxylic acids and isosteres of heterocyclic ring compounds having multiple heteroatoms for vision and memory disorders
US6303609B1 (en) * 1998-11-18 2001-10-16 Dupont Pharmaceuticals Company Isoxazoline fibrinogen receptor antagonists
WO2004105750A1 (en) * 2003-05-30 2004-12-09 Neuromed Technologies, Inc. 3-aminomethyl-pyrrolidines as n-type calcium channel blockers
US7244758B2 (en) 2003-05-30 2007-07-17 Neuromed Pharmaceuticals Ltd. N-type calcium channel blockers
US7388008B2 (en) 2004-08-02 2008-06-17 Ambrilia Biopharma Inc. Lysine based compounds
US8008297B2 (en) 2004-08-02 2011-08-30 Ambrilia Biopharma Inc. Lysine based compounds
WO2006114001A1 (en) * 2005-04-27 2006-11-02 Ambrilia Biopharma Inc. Method for improving pharmacokinetics of protease inhibitors and protease inhibitor precursors
JP2008543841A (en) * 2005-06-14 2008-12-04 シェーリング コーポレイション Macrocyclic heterocyclic aspartyl protease inhibitors
US8227450B2 (en) 2005-11-30 2012-07-24 Ambrilia Biopharma Inc. Lysine-based prodrugs of aspartyl protease inhibitors and processes for their preparation
US8580995B2 (en) 2005-11-30 2013-11-12 Taimed Biologics, Inc. Lysine-based prodrugs of aspartyl protease inhibitors and processes for their preparation
US7803827B2 (en) 2006-06-20 2010-09-28 Wyeth Llc Kv1.5 potassium channel inhibitors
US7504517B2 (en) 2006-06-20 2009-03-17 Wyeth Kv1.5 potassium channel inhibitors
US8410300B2 (en) 2006-09-21 2013-04-02 Taimed Biologics, Inc. Protease inhibitors
US8742158B2 (en) 2006-09-21 2014-06-03 TaiMed Biologies, Inc. Protease inhibitors
US9701727B2 (en) 2011-06-29 2017-07-11 The Trustees Of Columbia University In The City Of New York Inhibitor of neuronal connectivity linked to schizophrenia susceptibility and cognitive dysfunction
CN103626713A (en) * 2012-08-29 2014-03-12 中国科学院上海药物研究所 Aza (oxa)-cyclopentane-2- ketone (thione) compounds, medicine compositions thereof, preparation method and applications
CN105111198A (en) * 2015-08-05 2015-12-02 贵州大学 Nitryl isoxazole spliced 2-pyrrolidone compounds, and preparation method and application thereof
CN105111198B (en) * 2015-08-05 2018-08-07 贵州大学 Nitro isoxazole splices 2-Pyrrolidone class compound and preparation method and application

Also Published As

Publication number Publication date
PL328223A1 (en) 1999-01-18
CZ234498A3 (en) 1998-10-14
HUP9901024A2 (en) 1999-09-28
JP2000501111A (en) 2000-02-02
AU1758097A (en) 1997-08-20
NO983435L (en) 1998-09-21
AP844A (en) 2000-06-09
AU709239B2 (en) 1999-08-26
CA2243121A1 (en) 1997-07-31
EP0882022A1 (en) 1998-12-09
AP9700961A0 (en) 1997-04-30
BR9707086A (en) 1999-04-13
IL125479A0 (en) 1999-03-12
NO983435D0 (en) 1998-07-24

Similar Documents

Publication Publication Date Title
AU709239B2 (en) Aspartyl protease inhibitors
EP1136479B1 (en) Oxygenated-heterocycle containing sulfonamide inhibitors of aspartyl protease
EP0846110B1 (en) Thf-containing sulfonamide inhibitors of aspartyl protease
US9873690B2 (en) 3-indol substituted derivatives, pharmaceutical compositions and methods for use
JP4808380B2 (en) Carbamate esters as HIV protease inhibitors
SK9672000A3 (en) Prodrugs of aspartyl protease inhibitors
WO1990004917A1 (en) Heterocyclic peptide renin inhibitors
HUT74006A (en) Hiv protease inhibitors in pharmaceutical combinations and pharmaceutical compns. contg. them
MXPA97008055A (en) Consistent aspartile protease inhibitors ensulfonamide containing oxygen heterocicle

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE HU IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2243121

Country of ref document: CA

Ref document number: 2243121

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/1998/005970

Country of ref document: MX

Ref document number: PV1998-2344

Country of ref document: CZ

ENP Entry into the national phase

Ref document number: 1997 527124

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 199800661

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 1997904911

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV1998-2344

Country of ref document: CZ

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1997904911

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1997904911

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: PV1998-2344

Country of ref document: CZ