WO1999059989A1 - Process for the preparation of a benzo fused heterocyclo sulfonyl halide intermediate - Google Patents

Process for the preparation of a benzo fused heterocyclo sulfonyl halide intermediate Download PDF

Info

Publication number
WO1999059989A1
WO1999059989A1 PCT/US1999/007047 US9907047W WO9959989A1 WO 1999059989 A1 WO1999059989 A1 WO 1999059989A1 US 9907047 W US9907047 W US 9907047W WO 9959989 A1 WO9959989 A1 WO 9959989A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino
phenylmethyl
hydroxy
methylpropyl
mmol
Prior art date
Application number
PCT/US1999/007047
Other languages
French (fr)
Inventor
Sastry A. Kunda
Leo J. Letendre
Gary A. De Crescenzo
Original Assignee
G.D. Searle & Co.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by G.D. Searle & Co. filed Critical G.D. Searle & Co.
Priority to AU38604/99A priority Critical patent/AU3860499A/en
Publication of WO1999059989A1 publication Critical patent/WO1999059989A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/78Benzo [b] furans; Hydrogenated benzo [b] furans
    • C07D307/79Benzo [b] furans; Hydrogenated benzo [b] furans with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to carbon atoms of the hetero ring
    • 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
    • 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
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles 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 in position 2
    • C07D235/30Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/14Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D295/145Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/15Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/58Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/44Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D317/46Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D317/48Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring
    • C07D317/62Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring 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 atoms of the carbocyclic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D319/00Heterocyclic compounds containing six-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D319/101,4-Dioxanes; Hydrogenated 1,4-dioxanes
    • C07D319/141,4-Dioxanes; Hydrogenated 1,4-dioxanes condensed with carbocyclic rings or ring systems
    • C07D319/161,4-Dioxanes; Hydrogenated 1,4-dioxanes condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D319/18Ethylenedioxybenzenes, not substituted on the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D339/00Heterocyclic compounds containing rings having two sulfur atoms as the only ring hetero atoms
    • C07D339/02Five-membered rings
    • C07D339/06Five-membered rings having the hetero atoms in positions 1 and 3, e.g. cyclic dithiocarbonates
    • 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
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/0606Dipeptides with the first amino acid being neutral and aliphatic the side chain containing heteroatoms not provided for by C07K5/06086 - C07K5/06139, e.g. Ser, Met, Cys, Thr
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/0606Dipeptides with the first amino acid being neutral and aliphatic the side chain containing heteroatoms not provided for by C07K5/06086 - C07K5/06139, e.g. Ser, Met, Cys, Thr
    • C07K5/06069Ser-amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to retroviral 5 protease inhibitors and, more particularly, relates to novel compounds, composition and method for inhibiting retroviral proteases, such as human immunodeficiency virus (HIV) protease.
  • retroviral proteases such as human immunodeficiency virus (HIV) protease.
  • This invention in particular, relates to a ino acid hydroxyethylamine sulfonamide 10 protease inhibitor compounds, composition and method for inhibiting retroviral proteases, prophylactically preventing retroviral infection or the spread of a retrovirus, and treatment of a retroviral infection, e.g., an HIV infection.
  • the subject invention also 15 relates to processes for making such compounds as well as to intermediates useful in such processes.
  • gag and gag-pol gene transcription products are translated as proteins. These proteins are subsequently processed
  • gag precursor proteins are processed into the core proteins and the pol precursor proteins are processed into the viral enzymes, e.g.,
  • Retroviral protease inhibition typically involves a transition-state mimetic whereby the retroviral protease is exposed to a mimetic compound which binds (typically in a reversible manner) to the enzyme in competition with the gag and gag-pol proteins to thereby inhibit specific processing of structural proteins and the release of retroviral protease itself. In this manner, retroviral replication proteases can be effectively inhibited.
  • the present invention relates to selected retroviral protease inhibitor compounds, analogs and pharmaceutically acceptable salts, esters and prodrugs thereof.
  • the subject compounds are characterized as amino acid hydroxyethylamine sulfonamide inhibitor compounds.
  • the invention compounds advantageously inhibit retroviral proteases, such as human immunodeficiency virus (HIV) protease. Therefore, this invention also encompasses pharmaceutical compositions, methods for inhibiting retroviral proteases and methods for treatment or prophylaxis of a retroviral infection, such as an HIV infection.
  • the subject invention also relates to processes for making such compounds as well as to intermediates useful in such processes.
  • n 1 or 2;
  • R 1 represents alkyl, alkenyl, alkynyl, hydroxyalkyl, alkoxyalkyl, cyanoalkyl, imidazolylmethyl, -CH 2 CONH 2 , -CH 2 CH 2 CONH 2 , -CH 2 S(0) 2 NH 2 -CH 2 SCH 3 , -CH 2 S(0)CH 3 , -CH 2 S(0) 2 CH 3 , -C(CH 3 ) 2 SCH 3 , -C (CH 3 ) 2 S (0) CH 3 or -(CH 3 ) 2 S(0) 2 CH 3 radicals; preferably, R 1 represents alkyl of 1-5 carbon atoms, alkenyl of 2-5 carbon atoms, alkynyl of 2-5 carbon atoms, hydroxyalkyl of 1-3 carbon atoms, alkoxyalkyl of 1-3 alkyl and 1-3 alkoxy carbon atoms, cyanoalkyl of 1-3 alkyl carbon atoms, imidazolylmethyl, -
  • R 2 represents alkyl, aralkyl, alkylthioalkyl, arylthioalkyl or cycloalkylalkyl radicals; preferably,
  • R represents radicals of alkyl of 1-5 carbon atoms, aralkyl of 1-3 alkyl carbon atoms, alkylthioalkyl of 1-
  • R represents radicals of alkyl of 3-5 carbon atoms, arylmethyl, alkylthioalkyl of 1-3 alkyl carbon atoms, arylthiomethyl or cycloalkylmethyl of 5-6 ring member carbon atoms radicals; even more preferably, R represents isobutyl, n-butyl, CH 3 SCH 2 CH 2 -, benzyl, phenylthiomethyl, (2-naphthylthio)methyl, 4-methoxy phenylmethyl, 4-hydroxyphenylmethyl, 4- fluorophenylmethyl or cyclohexylmethyl radicals; even more preferably, R 2 represents benzyl, 4- fluorophenylmethyl or cyclohexylmethyl radicals; even more preferably, R 2 represents benzyl, 4- fluorophenylmethyl or cyclohexylmethyl
  • R 3 represents alkyl, cycloalkyl or cycloalkylalkyl radicals; preferably, R 3 represents radicals of alkyl radical of 1-5 carbon atoms, cycloalkyl of 5-8 ring members or cycloalkylmethyl radical of 3-6 ring members; more preferably, R 3 represents propyl, isoamyl, isobutyl, butyl, cyclopentylmethyl, cyclohexylmethyl, cyclohexyl or cycloheptyl radicals; more preferably R 3 represents isobutyl or cyclopentylmethyl radicals; R 4 represents aryl, heteroaryl or heterocyclo radicals; preferably, R represents aryl, benzo fused 5 to 6 ring member heteroaryl or benzo fused 5 to 6 ring member heterocyclo radicals; or
  • R 4 represents a radical of the formula
  • a and B each independently represent 0, S, SO or S0 ; preferably, A and B each represent 0;
  • R 6 represents hydrogen deuterium, alkyl or halogen radicals; preferably, R 6 represents hydrogen deuterium, alkyl of 1-5 carbon atoms, fluoro or chloro radicals; more preferably R 6 represents hydrogen deuterium, methyl, ethyl, propyl, isopropyl or fluoro radicals;
  • R 7 represents hydrogen, deuterium, alkyl or halogen radicals; preferably, R 7 represents hydrogen, deuterium, alkyl of 1-3 carbon atoms, fluoro or chloro radicals; more preferably, R 7 represents hydrogen, deuterium, methyl or fluoro radicals; or R 6 and R 7 each independently represent fluoro or chloro radicals; and preferably, R 6 and R 7 each represent a fluoro radical; or
  • R represents a radical of the formula
  • R 9 represents a radical of formula
  • Y represents 0, S or NH
  • X represents a bond, 0 or NR 21 ;
  • R 20 represents hydrogen, alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, heterocycloalkyl, a inoalkyl, N- mono-substituted or N,N-disubstituted aminoalkyl wherein said substituents are alkyl or aralkyl radicals, carboxyalkyl, alkoxycarbonylalkyl, cyanoalkyl or hydroxyalkyl radicals; preferably, R 20 represents hydrogen, alkyl of 1 to 5 carbon atoms, alkenyl of 2 to 5 carbon atoms, alkynyl of 2 to 5 carbon atoms, aralkyl of 1 to 5 alkyl carbon atoms, heteroaralkyl of 5 to 6 ring members and 1 to 5 alkyl carbon atoms, heterocycloalkyl of 5 to 6 ring members and 1 to 5 alkyl carbon atoms, aminoalkyl of 2 to 5 carbon atoms, N-mono- substitute
  • R 21 represents hydrogen or alkyl radicals; preferably, R 21 represents hydrogen radical or alkyl radical of 1 to 3 carbon atoms; more preferably, R 21 represents hydrogen or methyl radicals; and most preferably, R 21 represents a hydrogen radical; or
  • the radical of formula -NR 20 R 21 represents a heterocyclo radical; preferably, the radical of formula -NR 20 R 21 represents a 5 to 6 ring member heterocyclo radical; more preferably, the radical of formula -NR 20 R 21 represents pyrrolidinyl, piperidinyl, piperazinyl, 4- methylpiperazinyl, 4-benzylpiperazinyl, morpholinyl or thiamorpholinyl radicals; and
  • R 22 represents alkyl or R 20 R 21 N-alkyl radicals; preferably, R 22 represents alkyl or R 20 R 21 N-alkyl radicals wherein alkyl is 1 to 3 carbon atoms; and more preferably, R 22 represents alkyl radical of 1 to 3 carbon atoms; and
  • R 4 represents phenyl, 2-naphthyl, 4- methoxyphenyl , 4-hydroxyphenyl, 3 ,4-dimethoxyphenyl, 3- aminophenyl, 4-aminophenyl, benzothiazol-5-yl, benzothiazol-6-yl, 2-amino-benzothiazol-5-yl, 2- (methoxycarbonylamino)benzothiazol-5-yl, 2-amino- benzothiazol-6-yl, 2- (methoxycarbonylamino) benzothiazol-6-yl, 5-benzoxazolyl, 6-benzoxazolyl, 6- benzopyranyl, 3 , 4-dihydrobenzopyran-6-yl, 7- benzopyranyl, 3 , 4-dihydrobenzopyran-7-yl, 2,3- dihydrobenzofuran-5-yl, benzofuran-5-yl, 1,3- benzodioxol-5-
  • R 4 represents phenyl, 4-methoxyphenyl, 4-hydroxyphenyl, benzothiazol-5-yl, benzothiazol-6-yl, 2,3-dihydrobenzofuran-5-yl, benzofuran-5-yl, 1,3- benzodioxol-5-yl, 2-methyl-l, 3-benzodioxol-5-yl, 2,2- dimethyl-1, 3-benzodioxol-5-yl, 2 , 2-dideutero-l, 3- benzodioxol-5-yl, 2 , 2-difluoro-1, 3-benzodioxol-5-yl, l,4-benzodioxan-6-yl, 2- (methoxycarbonylamino) benzothiazol-6-
  • R 10 represents hydrogen, alkyl, hydroxyalkyl or alkoxyalkyl radicals, wherein alkyl is 1-3 carbon atoms; preferably R represents a hydrogen radical;
  • R represents alkyl radical of 1-5 carbon atoms, preferably methyl, isopropyl, butyl, secbutyl or isobutyl radicals; hydroxyalkyl radical of 1-4 carbon atoms, preferably hydroxymethyl or hydroxyethyl radicals; alkoxyalkyl radical of 1-4 alkyl carbon atoms, preferably methoxymethyl or methoxyethyl radicals; or hydrogen, benzyl, imidazolylmethyl, -CH 2 CH 2 CONH 2 , -CH 2 CONH 2 , -CH 2 CH 2 SCH 3 or -CHSCH 3 radicals or the sulfone or sulfoxide derivatives thereof; more preferably R 11 represents a hydrogen radical;
  • R 12 represents hydrogen, hydroxyalkyl or alkoxyalkyl radicals; preferably, R 12 represents hydrogen, hydroxyalkyl or alkoxyalkyl radicals, wherein alkyl is 1-3 carbon atoms; preferably R 12 represents a hydrogen radical; and
  • R 13 and R 14 each independently represent hydrogen, hydroxy, alkoxy, hydroxyalkoxy, hydroxyalkyl or alkoxyalkyl radicals; preferably, R 13 and R each independently represent hydrogen, hydroxy, alkoxy, 2- hydroxyethoxy, hydroxyalkyl or alkoxyalkyl radicals, wherein alkyl is 1-3 carbon atoms; more preferably, R and R 14 each independently represent hydrogen, hydroxy, methoxy or ethoxy radicals; or
  • R 12 and R 13 or R 13 and R 14 along with the carbon atoms to which they are attached represent 5-6 ring membered heteroaryl or benzo radical, each of which is optionally substituted with at least one hydroxy or alkoxy radical of 1-3 carbon atoms; preferably R 12 and R 13 or R 13 and R 14 along with the carbon atoms to which they are attached represent benzo radical, which is optionally substituted with at least one hydroxy or methoxy radical.
  • the absolute stereochemistry of the carbon atom of -CH(OH)- group is preferably (R) .
  • the absolute stereochemistry of the carbon atom of -CH(R 1 )- group is preferably (S) .
  • the absolute stereochemistry of the carbon atom of-CH(R 2 )- groups is preferably (S) .
  • a family of compounds of particular interest within Formula I are compounds embraced by the formula
  • n R 1 , R 2 , R 3 , and R 4 are as defined above.
  • a family of compounds of further interest within Formula II are compounds embraced by the formula
  • R 1 , R 2 , R 3 , R 4 and n are as defined above.
  • a more preferred family of compounds within Formula III consists of compounds or a pharmaceutically acceptable salt, prodrug or ester thereof, wherein n represents 1;
  • R 1 represents sec-butyl, tert-butyl, iso-propyl, 3- propynyl or -C(CH 3 ) 2 S(0) 2 CH 3 radicals;
  • R 2 represents a benzyl radical;
  • R 3 represents propyl, isoa yl, isobutyl, butyl, cyclohexyl, cycloheptyl, cyclopentylmethyl or cyclohexylmethyl radicals;
  • R 4 is as defined above
  • Compounds of interest include the following:
  • alkyl means a straight-chain or branched-chain alkyl radical containing preferably from 1 to 8 carbon atoms, more preferably from 1 to 5 carbon atoms, most preferably 1 to 3 carbon atoms.
  • examples of such radicals include methyl, ethyl, n-propyl, isopropyl, n- butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso- amyl, hexyl, octyl and the like.
  • alkenyl alone or in combination, means a straight-chain or branched-chain hydrocarbon radical having one or more double bonds and containing preferably from 2 to 10 carbon atoms, more preferably from 2 to 8 carbon atoms, most preferably from 2 to 5 carbon atoms.
  • suitable alkenyl radicals include ethenyl, propenyl, 2- methylpropenyl, 1,4-butadienyl and the like.
  • alkynyl alone or in combination, means a straight- chain or branched chain hydrocarbon radical having one or more triple bonds and containing preferably from 2 to 10 carbon atoms, more preferably from 2 to 5 carbon atoms.
  • alkynyl radicals include ethynyl, propynyl (propargyl) , butynyl and the like.
  • alkoxy alone or in combination, means an alkyl ether radical wherein the term alkyl is as defined above.
  • suitable alkyl ether radicals include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso- butoxy, sec-butoxy, tert-butoxy and the like.
  • cycloalkyl alone or in combination, means a saturated or partially saturated monocyclic, bicyclic or tricyclic alkyl radical wherein each cyclic moiety contains preferably from 3 to 8 carbon atom ring members, more preferably from 3 to 7 carbon atom ring members, most preferably from 5 to 6 carbon atom ring members, and which may optionally be a benzo fused ring system which is optionally substituted as defined herein with respect to the definition of aryl.
  • cycloalkyl radicals examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, octahydronaphthyl, 2,3-dihydro- lH-indenyl, ada antyl and the like.
  • "Bicyclic” and “tricyclic” as used herein are intended to include both fused ring systems, such as naphthyl and ⁇ -carbolinyl, and substituted ring systems, such as biphenyl, phenylpyridyl, naphthyl and diphenylpiperazinyl.
  • cycloalkylalkyl means an alkyl radical as defined above which is substituted by a cycloalkyl radical as defined above.
  • examples of such cycloalkylalkyl radicals include cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, 1-cyclopentylethyl, 1-cyclohexylethyl, 2-cyclopentylethyl, 2- cyclohexylethyl, cyclobutylpropyl, cyclopentylpropyl, cyclohexylbutyl and the like.
  • aryl alone or in combination, means a phenyl or naphthyl radical which is optionally substituted with one or more substituents selected from alkyl, alkoxy, halogen, hydroxy, amino, nitro, cyano, haloalkyl, carboxy, alkoxycarbonyl, cycloalkyl, heterocyclo, alkanoylamino, amido, amidino, alkoxycarbonylamino, N-alkylamidino, alkylamino, dialkylamino, N-alkylamido, N,N-dialkylamido, aralkoxycarbonylamino, alkylthio, alkylsulfinyl, alkylsulfonyl and the like.
  • aryl radicals are phenyl, p-tolyl, 4-methoxyphenyl, 4-(tert- butoxy) phenyl, 3-methyl-4-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 3-nitrophenyl, 3-aminophenyl, 3- acetamidophenyl, 4-acetamidophenyl, 2-methyl-3- acetamidophenyl, 4-CF 3 -phenyl, 2-methyl-3-aminophenyl, 3-methy1-4-aminopheny1, 2-amino-3- methylphenyl , 2,4-dimethyl-3-aminophenyl, 4- hydroxyphenyl, 3-methyl-4-hydroxyphenyl, 1-naphthyl, 2- naphthyl, 3-amino-l-naphthyl, 2-methyl-3-amino-l- naphthyl, 6-amino-2-naphthyl, 4
  • aralkyl and “aralkoxy”, alone or in combination, means an alkyl or alkoxy radical as defined above in which at least one hydrogen atom is replaced by an aryl radical as defined above, such as benzyl, benzyloxy, 2-phenylethyl, dibenzylmethyl, hydroxyphenylmethyl, methylphenylmethy1 , diphenylmethyl, diphenylmethoxy, 4-methoxyphenylmethoxy and the like.
  • aralkoxycarbonyl alone or in combination, means a radical of the formula aralkyl-O- C(0)- in which the term “aralkyl” has the significance given above.
  • aralkoxycarbonyl radical examples include benzyloxycarbonyl and 4- methoxyphenylmethoxycarbonyl .
  • aryloxy means a radical of the formula aryl-O- in which the term aryl has the significance given above.
  • alkanoyl alone or in combination, means an acyl radical derived from an alkanecarboxylic acid, examples of which include acetyl, propionyl, butyryl, valeryl, 4-methylvaleryl, and the like.
  • cycloalkylcarbonyl means an acyl radical of the formula cycloalkyl-C(O) - in which the term “cycloalkyl” has the significance give above, such as cyclopropylcarbonyl, cyclohexylcarbonyl, adamantylcarbonyl, 1, 2 , 3 ,4-tetrahydro-2-naphthoyl, 2- acetamido-1, 2,3, 4-tetrahydro-2-naphthoyl, 1-hydroxy- l,2,3,4-tetrahydro-6-naphthoyl and the like.
  • cycloalkylcarbonyl means an acyl radical of the formula cycloalkyl-C(O) - in which the term “cycloalkyl” has the significance give above, such as cyclopropylcarbonyl, cyclohexylcarbonyl, adamantylcarbonyl, 1, 2 , 3 ,4
  • aralkanoyl means an acyl radical derived from an aryl- substituted alkanecarboxylic acid such as phenylacetyl, 3-phenylpropionyl (hydrocinnamoyl) , 4-phenylbutyryl, (2- naphthyl) acetyl, 4-chlorohydrocinnamoyl, 4- aminohydrocinnamoyl, 4-methoxyhydrocinnamoyl, and the like.
  • the term “aroyl” means an acyl radical derived from an arylcarboxylic acid, "aryl” having the meaning given above. Examples of such aroyl radicals include substituted and unsubstituted benzoyl or napthoyl such as benzoyl, 4-chlorobenzoyl, 4-carboxybenzoyl, 4-
  • heterocyclo means a saturated or partially unsaturated monocyclic, bicyclic or tricyclic heterocycle radical containing at least one, preferably 1 to 4 , more preferably 1 to 2, nitrogen, oxygen or sulfur atom ring members and having preferably 3 to 8 ring members in each ring, more preferably 3 to 7 ring members in each ring and most preferably 5 to 6 ring members in each ring.
  • Heterocyclo is intended to include sulfones, sulfoxides, N-oxides of tertiary nitrogen ring members, and carbocyclic fused and benzo fused ring systems.
  • a secondary nitrogen atom i.e., -NH-
  • Heterocycloalkyl means an alkyl radical as defined above in which at least one hydrogen atom is replaced by a heterocyclo radical as defined above, such as pyrrolidinylmethy1, tetrahydrothienyl ethyl, pyridylmethyl and the like.
  • heteroaryl alone or in combination, means an aromatic heterocyclo radical as defined above, which is optionally substituted as defined above with respect to the definitions of aryl and heterocyclo.
  • heterocyclo and heteroaryl groups examples include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiamorpholinyl, pyrrolyl, imidazolyl (e.g., imidazol 4-yl, 1- benzyloxycarbonylimidazol-4-yl, etc.), pyrazolyl, pyridyl, (e.g., 2- (l-piperidinyl)pyridyl and 2-(4-benzyl piperazin-1-yl-l-pyridinyl, etc.), pyrazinyl, pyrimidinyl, furyl, tetrahydrofuryl, thienyl, tetrahydrothienyl and its sulfoxide and sulfone derivatives, triazolyl, oxazolyl, thiazolyl, indolyl (e.g., 2-indolyl, etc.), quinolinyl,
  • cycloalkylalkoxycarbonyl means an acyl group derived from a cycloalkylalkoxycarboxylic acid of the formula cycloalkylalkyl-O-COOH wherein cycloalkylalkyl has the meaning given above.
  • aryloxyalkanoyl means an acyl radical of the formula aryl-O-alkanoyl wherein aryl and alkanoyl have the meaning given above.
  • heterocycloalkoxycarbonyl means an acyl group derived from heterocycloalkyl-O-COOH wherein heterocycloalkyl is as defined above.
  • heterocycloalkanoyl is an acyl radical derived from a heterocycloalkylcarboxylic acid wherein heterocyclo has the meaning given above.
  • heterocycloalkoxycarbonyl means an acyl radical derived from a heterocycloalkyl-O-COOH wherein heterocyclo has the meaning given above.
  • heteroaryloxycarbonyl means an acyl radical derived from a carboxylic acid represented by heteroaryl-O-COOH wherein heteroaryl has the meaning given above.
  • a inocarbonyl alone or in combination, means an amino-substituted carbonyl (carbamoyl) group wherein the amino group can be a primary, secondary or tertiary amino group containing substituents selected from alkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl radicals and the like.
  • aminoalkanoyl means an acyl group derived from an amino-substituted alkylcarboxylic acid wherein the amino group can be a primary, secondary or tertiary amino group containing substituents selected from alkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl radicals and the like.
  • halogen means fluorine, chlorine, bromine or iodine.
  • haloalkyl means an alkyl radical having the meaning as defined above wherein one or more hydrogens are replaced with a halogen.
  • haloalkyl radicals examples include chloromethyl, 1-bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1, 1, 1-trifluoroethyl and the like.
  • the term "leaving group" generally refers to groups readily displaceable by a nucheophile(such as an a ine, a thiol or an alcohol nucleophile. Such leaving groups are well known in the art. Examples of such leaving groups include, but are not limited to, N-hydroxysuccinimide, N- hydroxybenzotriazole, halides, triflates, tosylates and the like. Preferred leaving groups are indicated herein where appropriate.
  • Procedures for preparing the compounds of Formula I are set forth below. It should be noted that the general procedure is shown as it relates to preparation of compounds having the specified stereochemistry, for example, wherein the absolute stereochemistry about the hydroxyl group is designated as (R) . However, such procedures are generally applicable to those compounds of opposite configuration, e.g., where the stereochemistry about the hydroxyl group is (S) .
  • the compounds having the (R) stereochemistry can be utilized to produce those having the (S) stereochemistry.
  • a compound having the (R) stereochemistry can be inverted to the (S) stereochemistry using well-known methods.
  • P represents an amino protecting group
  • R 2 is as defined above
  • Suitable amino protecting groups are well known in the art and include carbobenzoxy, t-butoxycarbonyl, and the like.
  • a preferred amino protecting group is carbobenzoxy.
  • a preferred N-protected chloroketone is N-benzyloxycarbonyl-L-phenylalanine chloromethyl ketone.
  • a preferred reducing agent is sodium borohydride.
  • the reduction reaction is conducted at a temperature of from -10°C to about 25°C, preferably at about 0°C, in a suitable solvent system such as, for example, tetrahydrofuran, and the like.
  • the N-protected chloroketones are commercially available, e.g., such as from Bachem, Inc., Torrance, California.
  • the chloroketones can be prepared by the procedure set forth in S. J. Fittkau, J. Prakt. Chem. , 315, 1037 (1973) , and subsequently N-protected utilizing procedures which are well known in the art.
  • the halo alcohol can be utilized directly, as described below, or, preferably, is reacted, preferably at room temperature, with a suitable base in a suitable solvent system to produce an N-protected amino epoxide of the formula:
  • Suitable solvent systems for preparing the amino epoxide include ethanol, methanol, isopropanol, tetrahydrofuran, dioxane, and the like including mixtures thereof.
  • Suitable bases for producing the epoxide from the reduced chloroketone include potassium hydroxide, sodium hydroxide, potassium t-butoxide, DBU and the like. A preferred base is potassium hydroxide.
  • a protected amino epoxide can be prepared, such as in co-owned and co-pending PCT Patent Application Serial No. PCT/US93/04804 (WO 93/23388) and PCT/US94/12201, and US Patent Application Attorney
  • P represents carboxyl-protecting group, e.g., methyl, ethyl, benzyl, tertiary-butyl, 4- methoxyphenylmethy1 and the like;
  • R 2 is as defined above; and
  • P 1 and P2 i•ndependently are selected from amine protecting groups, including but not limited to, aralkyl, substituted aralkyl, cycloalkenylalkyl and substituted cycloalkenylaloyl, allyl, substituted allyl, acyl, alkoxycarbonyl, aralkoxycarbonyl and silyl.
  • aralkyl examples include, but are not limited to benzyl, ortho-methylbenzyl, trityl and benzhydryl, which can be optionally substituted with halogen, alkyl of C ⁇ Cg, alkoxy, hydroxy, nitro, alkylene, amino, alkylamino, acylamino and acyl, or their salts, such as phosphonium and ammonium salts.
  • aryl groups include phenyl, naphthalenyl, indanyl, anthracenyl, durenyl, 9- (9-phenylfluorenyl) and phenanthrenyl, cycloalkenylalkyl or substituted cycloalkylenylalkyl radicals containing cycloalkyls of Cg-Cio- Suitable acyl groups include carbobenzoxy, t-butoxycarbonyl, iso-butoxycarbonyl, benzoyl, substituted benzoyl, butyryl, acetyl, tri- fluoroacetyl, tri-chloroacetyl, phthaloyl and the like.
  • P 1 and P 2 are independently selected from aralkyl and substituted aralkyl. More preferably, each of P 1 and P 2 is benzyl.
  • the P 1 and/or P 2 protecting groups can form a heterocyclic ring with the nitrogen to which they are attached, for example, 1,2- bis(methylene) benzene, phthali idyl, succinimidyl, maleimidyl and the like and where these heterocyclic groups can further include adjoining aryl and cycloalkyl rings.
  • the heterocyclic groups can be mono-, di- or tri-substituted, e.g., nitrophthalimidyl.
  • silyl refers to a silicon atom optionally substituted by one or more alkyl, aryl and aralkyl groups.
  • Suitable silyl protecting groups include, but are not limited to, trimethylsilyl, triethylsilyl, tri- isopropylsilyl , tert-butyldimethylsilyl, dimethylphenylsilyl, 1,2-bis (dimethylsilyl) benzene, 1,2- bis(dimethylsilyl) ethane and diphenylmethylsilyl.
  • Silylation of the amine functions to provide mono- or bis-disilylamine can provide derivatives, of the aminoalcohol, amino acid, amino acid esters and amino acid amide. In the case of amino acids, amino acid esters and amino acid amides, reduction of the carbonyl function provides the required mono- or bis-silyl aminoalcohol.
  • Silylation of the aminoalcohol can lead to the N,N,0-tri-silyl derivative.
  • Removal of the silyl function from the silyl ether function is readily accomplished by treatment with, for example, a metal hydroxide or ammonium flouride reagent, either as a discrete reaction step or in situ during the preparation of the amino aldehyde reagent.
  • Suitable silylating agents are, for example, trimethylsilyl chloride, tert- buty-dimethylsilyl chloride, phenyldimethylsilyl chloride, diphenylmethylsilyl chloride or their combination products with imidazole or DMF.
  • Methods for silylation of amines and removal of silyl protecting groups are well known to those skilled in the art. Methods of preparation of these amine derivatives from corresponding amino acids, amino acid amides or amino acid esters are also well known to those skilled in the art of organic chemistry including amino acid/amino acid ester or aminoalcohol
  • the amino-protected L-amino acid ester is then reduced, to the corresponding alcohol.
  • the amino-protected L-amino acid ester can be reduced with diisobutylaluminum hydride at -78 °C in a suitable solvent such as toluene.
  • Preferred reducing agents include lithium aluminium hydride, lithium borohydride, sodium borohydride, borane, lithium tri-ter- butoxyaluminum hydride, borane/THF complex.
  • the reducing agent is diisobutylaluminum hydride (DiBAL-H) in toluene.
  • the resulting alcohol is then converted, for example, by way of a Swern oxidation, to the corresponding aldehyde, of the formula:
  • Acceptable oxidizing reagents include, for example, sulfur trioxide-pyridine complex and DMSO, oxalyl chloride and DMSO, acetyl chloride or anhydride and DMSO, trifluoroacetyl chloride or anhydride and DMSO, methanesulfonyl chloride and DMSO or tetrahydro thiaphene-S-oxide, toluenesulfonyl bromide and DMSO, trifluoromethanesulfonyl anhydride (triflic anhydride) and DMSO, phosphorus pentachloride and DMSO, dimethylphosphoryl chloride and DMSO and isobutyl chloroformate and DMSO.
  • the oxidation conditions reported by Reetz et al (An ew Che . , 99, p. 1186,
  • the reaction may be carried out under an inert atmosphere such as nitrogen or argon, or normal or dry air, under atmospheric pressure or in a sealed reaction vessel under positive pressure.
  • an inert atmosphere such as nitrogen or argon, or normal or dry air
  • Preferred is a nitrogen atmosphere.
  • Alternative amine bases include, for example, tri-butyl amine, tri-isopropyl amine, N- methylpiperidine, N-methyl morpholine, azabicyclononane, diisopropylethylamine, 2,2,6, 6-tetramethylpiperidine, N,N-dimethylaminopyridine, or mixtures of these bases.
  • Triethylamine is a preferred base.
  • DMSO dimethylformamide
  • dimethylacetamide dimethylacetamide
  • acetamide tetramethyl urea and its cyclic analog
  • N-methylpyrrolidone sulfolane and the like.
  • N,N-dibenzylphenylalaninol as the aldehyde precursor
  • hydride reduction of an amide or ester derivative of the corresponding benzyl (or other suitable protecting group) nitrogen protected phenylalanme, substituted phenylalanme or cycloalkyl analog of phenylalanine derivative can be carried out to provide the aldehydes. Hydride transfer is an additional method of aldehyde synthesis under conditions where aldehyde condensations are avoided, cf, Oppenauer Oxidation.
  • the aldehydes of this process can also be prepared by methods of reducing protected phenylalanine and phenylalanine analogs or their amide or ester derivatives by, e.g., sodium amalgam with HC1 in ethanol or lithium or sodium or potassium or calcium in ammonia.
  • the reaction temperature may be from about -20°C to about 45°C, and preferably from abut 5°C to about 25°C.
  • Two additional methods of obtaining the nitrogen protected aldehyde include oxidation of the corresponding alcohol with bleach in the presence of a catalytic amount of 2 , 2, 6, 6-tetramethyl-l-pyridyloxy free radical.
  • oxidation of the alcohol to the aldehyde is accomplished by a catalytic amount of tetrapropylammonium perruthenate in the presence of N-methylmorpholine-N-oxide.
  • an acid chloride derivative of a protected phenylalanine or phenylalanine derivative as disclosed above can be reduced with hydrogen and a catalyst such as Pd on barium carbonate or barium sulphate, with or without an additional catalyst moderating agent such as sulfur or a thiol (Rosenmund Reduction) .
  • a catalyst such as Pd on barium carbonate or barium sulphate
  • an additional catalyst moderating agent such as sulfur or a thiol (Rosenmund Reduction)
  • the aldehyde resulting from the Swern oxidation is then reacted with a halomethyllithium reagent, which reagent is generated in situ by reacting an alkyllithium or arylithium compound with a dihalomethane represented by the formula X 1 CH 2 X 2 wherein X 1 and X 2 independently represent I, Br or Cl.
  • the diastereomers can be separated e.g., by chromatography, or, alternatively, once reacted in subsequent steps the diastereomeric products can be separated.
  • a D-amino acid can be utilized in place of the L-amino acid in order to prepare compounds having an (S) stereochemistry at the carbon bonded to R .
  • chloromethylithium or bromomethylithium to a chiral amino aldehyde is highly diastereoselective.
  • the chloromethyllithium or bromomethylithium is generated in-situ from the reaction of the dihalomethane and n-butyllithium.
  • Acceptable methyleneating halomethanes include chloroiodomethane, bromochloromethane, dibromomethane, diiodomethane, bromofluoromethane and the like.
  • the sulfonate ester of the addition product of, for example, hydrogen bromide to formaldehyde is also a methyleneating agent.
  • Tetrahydrofuran is the preferred solvent, however alternative solvents such as toluene, dimethoxyethane, ethylene dichloride, methylene chloride can be used as pure solvents or as a mixture.
  • Dipolar aprotic solvents such as acetonitrile, DMF, N- methylpyrrolidone are useful as solvents or as part of a solvent mixture.
  • the reaction can be carried out under an inert atmosphere such as nitrogen or argon.
  • organometalic reagents reagents such as methyllithium, tert-butyl lithium, sec-butyl lithium, phenyllithium, phenyl sodium and the like.
  • the reaction can be carried out at temperatures of between about -80°C to 0°C but preferably between about -80°C to -20°C.
  • the most preferred reaction temperatures are between -40°C to - 15°C.
  • Reagents can be added singly but multiple additions are preferred in certain conditions.
  • the preferred pressure of the reaction is atmospheric however a positive pressure is valuable under certain conditions such as a high humidity environment.
  • Alternative methods of conversion to the epoxides of this invention include substitution of other charged methylenation precurser species followed by their treatment with base to form the analogous anion.
  • these species include trimethylsulfoxonium tosylate or triflate, tetramethylammonium halide, methyldiphenylsulfoxonium halide wherein halide is chloride, bromide or iodide.
  • the conversion of the aldehydes of this invention into their epoxide derivative can also be carried out in multiple steps.
  • the protected aminosulfide alcohol can be alkylated with, for example, the alkylating agents above, to provide a sulfonium salts that are subsequently converted into the subject epoxides with tert-amine or mineral bases.
  • the desired epoxides formed using most preferred conditions, diastereoselectively in ratio amounts of at least about an 85:15 ratio (S:R) .
  • the product can be purified by chromatography to give the diastereomerically and enantiomerically pure product but it is more conveniently used directly without purification to prepare retroviral protease inhibitors.
  • the foregoing process is applicable to mixtures of optical isomers as well as resolved compounds.
  • a particular optical isomer can be selected by the choice of starting material, e.g., L- phenylalanine, D-phenylalanine, L-phenylalaninol, D- phenylalaninol, D-hexahydrophenylalaninol and the like, or resolution can occur at intermediate or final steps.
  • Chiral auxiliaries such as one or two equivalents of camphor sulfonic acid, citric acid, camphoric acid, 2- methoxyphenylacetic acid and the like can be used to form salts, esters or amides of the compounds of this invention.
  • the amino epoxide is then reacted, in a suitable solvent system, with an equal amount, or preferably an excess of, a desired amine of the formula R NH , wherein R is hydrogen or is as defined above.
  • the reaction can be conducted over a wide range of temperatures, e.g., from about 10°C to about 100°C, but is preferably, but not necessarily, conducted at a temperature at which the solvent begins to reflux.
  • Suitable solvent systems include protic, non-protic and dipolar aprotic organic solvents such as, for example, those wherein the solvent is an alcohol, such as methanol, ethanol, isopropanol, and the like, ethers such as tetrahydrofuran, dioxane and the like, and toluene, N,N-dimethylformamide, dimethyl sulfoxide, and mixtures thereof.
  • a preferred solvent is isopropanol.
  • the resulting product is a 3- (N-protected amino) -3-(R 2 ) -1-(NHR 3 ) -propan-2-ol derivative (hereinafter referred to as an amino alcohol) can be represented by the formulas:
  • P, P 1 , P 2 , R 2 and R 3 are as described above.
  • a haloalcohol can be utilized in place of the amino epoxide.
  • the amino alcohol defined above is then reacted in a suitable solvent with an activated sulfonyl function such as the sulfonyl chloride R 4 S0 2 C1, the sulfonyl bromide RS0 2 Br, the sulfonate ester or the corresponding sulfonyl anhydride, preferably in the presence of an acid scavenger.
  • an activated sulfonyl function such as the sulfonyl chloride R 4 S0 2 C1, the sulfonyl bromide RS0 2 Br, the sulfonate ester or the corresponding sulfonyl anhydride, preferably in the presence of an acid scavenger.
  • Suitable solvents in which the reaction can be conducted include methylene chloride, dichloromethane, ethyl acetate, tetrahydrofuran and the like.
  • Suitable acid scavengers include triethylamine, pyridine as well as hindered amine bases such as diazabicyclooctane, diazabicycloundecene, 2,6 disubstituted pyridine derivatives, di-isopropylamine and the like.
  • Inorganic bases such as sodium carbonate, potassium bicarbonate, calcium carbonate and the like may also be used if desired.
  • the process of preparing the sulfonamide can be carried out as a discrete step following isolation of, for example, a sulfonyl chloride reagent or as part of a single pot process during the preparation of the activated sulfonyl function. Isolation of the sulfonyl chloride is preferred.
  • the resulting sulfonamide derivative can be represented, depending on the epoxide utilized by the formulas
  • the sulfonyl halides of the formula R 4 S0 2 X can be prepared by the reaction of a suitable aryl, heteroaryl and benzo fused heterocyclo Grignard or lithium reagents with sulfuryl chloride, or sulfur dioxide followed by oxidation with a halogen, preferably chlorine.
  • Aryl, heteroaryl and benzo fused heterocyclo Grignard or lithium reagents can be prepared from their corresponding halide (such as chloro or bromo) compounds which are commercially available or readily prepared from commercially available starting materials using known methods in the art.
  • thiols may be oxidized to sulfonyl chlorides using chlorine in the presence of water under carefully controlled conditions.
  • sulfonic acids such as arylsulfonic acids, heteroaryl and benzo fused heterocyclosulfonic acids, may be converted into sulfonyl halides using reagents such as PC1 5 , S0C1 2 , C1C(0)C(0)C1 and the like, and also to anhydrides using suitable dehydrating reagents.
  • reagents such as PC1 5 , S0C1 2 , C1C(0)C(0)C1 and the like, and also to anhydrides using suitable dehydrating reagents.
  • the sulfonic acids may in turn be prepared using procedures well known in the art. Some sulfonic acids are commercially available.
  • sulfinyl halides RSOX
  • RSX sulfenyl halides
  • Arylsulfonic acids, benzo fused heterocyclo sulfonic acids or heteroaryl sulfonic acids can be prepared by sulfonation of the aromatic ring by well known methods in the art, such as by reaction with sulfuric acid, S0 3 , S0 3 complexes, such as DMF(S0 3 ) (also depicted as S0 3 DMF, S0 3 »DMF, DMFS0 3 and the like), pyridine (S0 3 ) , N,N-dimethylacetamide(S0 ) , and the like.
  • arylsulfonyl halides are prepared from aromatic compounds by reaction with DMF(S0 ) and S0C1 2 or
  • benzo fused heterocyclo sulfonyl halides are prepared from aromatic compounds by reaction with DMF(S0 3 ) and CIC(O) C(O) Cl.
  • the reactions may be performed step wise or in a single pot.
  • a single pot reaction is understood to mean a single reaction system, reaction vessel, kettle, bomb, flask, and the like.
  • amidation of the amine intermediate discussed above and used to prepare, for example, the sulfonamide may be carried out in either a one pot or two pot reaction or reaction series as indicated.
  • the reactions used in the method of converting a aromatic compound, including heteroaryl compounds and benzo fused heterocycles, into sulfonic,acids, activated sulfonic acids or sulfonamides is generally carried out under an inert atmosphere such as nitrogen, argon, or helium.
  • an inert atmosphere such as nitrogen, argon, or helium.
  • dry air is possible in some cases where it does not decrease the yield(s) or present a safety hazard.
  • the solvent or solvent mixtures are water immiscible, for example, dichloroethane (ethylene dichloride) , methylene chloride, trichloroethane, toluene, tetrachloroethane and the like.
  • a co-solvent may be used that is water miscible if desired.
  • the temperatures of the various stages of the process or processes can vary depending upon reagents and other conditions.
  • the process exemplified, for example, by the preparation of 1,3-benzodioxolesulfonyl chloride is started by mixing reagents at room temperature although a range of about
  • the S0 3 «DMF reagent is an especially desirable reagent allowing a range of reaction temperatures that can be controlled by rationing the amount of heat provided to the reaction vessel or the reflux temperature of the solvent(s).
  • the reaction can be worked up by methods know in the art, e.g., cooling, addition of water and/or an acid such as hydrochloric acid or sulfuric acid, to provide the sulfonic acid.
  • an intermediate such as the sulfonyl chloride by cooling the reaction mixture to a temperature of between about -25°C to about 65°C.
  • the preferred range is between about 0°C and room temperature.
  • the amount of cooling that is necessary will depend on the type of oxalyl halide that is added in the next step.
  • an oxalyl halide such as oxalyl chloride or oxalyl bromide can be added to the reaction vessel at such a rate as to control the reaction temperature.
  • the temperature can range from about 0°C to about 50°C but preferably between about 0°C and room temperature. Most preferred is a range of between about 10°C and about 40°C.
  • the reaction can be sampled for analysis. When analysis indicates that the conversion has been carried out, the excess reagents, if present, can be decomposed by the controlled addition of water. Care, such as that which would be expected of a trained person, is indicated in this operation. Isolation of the sulfonyl chloride proceeds by separation of the phases, washing, if desired, the aqueous phase with a water non-miscible solvent such as dichloroethane one to four times and drying the organic phases separately, in batches or as a single, combined solution.
  • a water non-miscible solvent such as dichloroethane
  • the drying agent can be a chemical such as magnesium sulfate or sodium sulfate or the water can be removed via an azeotrope.
  • the solution remaining, which may or may not be mixed with various amounts of drying agent, may be filtered if desired.
  • the solvent can be removed by distillation at atmospheric pressure or below or evaporation under reduced pressure using, for example, a rotary evaporator.
  • a crystallizing or washing process may provide product that is superior on an economic or scientific basis when used for the preparation of a intermediate substrate for catalytic processes such as reductive deprotection via removal of benzyl groups from an amine nitrogen.
  • the isolated activated sulfonic acid product may be added directly as a solid or dissolved in a solvent prior to addition as a reagent to the next reaction.
  • Isolation of, for example, a benzo fused heterocyclo sulfonyl chloride such as l,3-benzodioxole-5-sulfonyl chloride, prior to its reaction with an amine is preferred and the yield is high and may exceed 95%.
  • the conversion of the activated oxidized sulfur acid product into the sulfonamide, sulfinamide or sulfeneamide can be carried out in the same pot as its preparation if desired.
  • Addition of the amine following reaction, decomposition or removal of the oxalyl chloride reagent with or without the addition of a hydrogen ion (H + ) scavenger discussed above produces the amide product.
  • the same product is obtained when the reaction is partially worked up by the addition of water as presented above.
  • the activated sulfur acid such as the sulfonyl chloride discussed above is used to form the sulfonamide with or without the addition of a proton scavenging agent.
  • amine salt formed in this process can be isolated and reused according to the methods below. Should the product of, for example, the sulfonamide forming reaction is itself a basic amine, it may be isolated as a salt or as an amine.
  • Arylsulfonic acids, benzo fused heterocyclo sulfonic acids, heteroaryl sulfonic acids, arylmercaptans, benzo fused heterocyclo mercaptans, heteroarylmercaptans, arylhalides, benzo fused heterocyclo halides, heteroarylhalides, and the like are commercially available or can be readily prepared from starting materials commercially available using standard methods well known in the art.
  • a number of sulfonic acids R 4 S0 3 H represented by the formulas
  • A, B, Z, R 6 , R 7 and R 9 are as defined above, have been prepared from 1,2-benzenedithiol, 2- mercaptanphenol, 1, 2-benzenediol, 2-aminobenzothiazole, benzothiazole, 2-aminobenzimidazole, benzimidazole, and the like, which are commercially available, by Carter, US Patent 4,595,407; Ehrenfreund et al., US Patent 4,634,465; Yoder et al., J. Heterocycl. Chem. 4:166-167 (1967); Cole et al., Aust. J. Chem. 33:675-680 (1980); Cabiddu et al.
  • CF 2 Br 2 or CD 2 Br 2 can be reacted with 1, 2-benzenedithiol, 2-mercaptanphenol or 1,2- benzenediol in the presence of base to produce the compounds
  • a and B are O or S and D is a deuterium atom.
  • a and/or B represent S
  • the sulfur can be oxidized using the methods described below to the sulfone or sulfoxide derivatives.
  • the ami ⁇ no protecting group P or P1 and P2 amino protecting groups are removed under conditions which will not affect the remaining portion of the molecule.
  • These methods include acid hydrolysis, hydrogenolysis and the like.
  • a preferred method involves removal of the protecting group, e.g., removal of a carbobenzoxy group, by hydrogenolysis utilizing palladium on carbon in a suitable solvent system such as an alcohol, acetic acid, and the like or mixtures thereof.
  • the protecting group is a t-butoxycarbonyl group
  • it can be removed utilizing an inorganic or organic acid, e.g., HC1 or trifluoroacetic acid, in a suitable solvent system, e.g., dioxane or methylene chloride.
  • a suitable solvent system e.g., dioxane or methylene chloride.
  • the resulting product is the amine salt derivative.
  • the amine is then coupled to the DL-, D-, or L-amino acid corresponding to the formula PNHCH(R 1 )C00H, wherein P and R 1 are as defined above, followed by deprotection of the amine as described above, and coupling to
  • W is a leaving group, such as mesylate, bromo or chloro
  • L is leaving group such as halide, anhydride, active ester, and the like.
  • R 10 and R 11 are both hydrogen radical, bromoacetyl halide, chloroacetyl halide or the corresponding anhydride can be used.
  • pyrrolidine 2- pyrrolidinemethanol, 3-pyrrolidinol, 2- (methoxymethyl) pyrrolidine, kainic acid, piperidine, 3- hydroxypiperidine, 4-hydroxypiperidine, 2- piperidinemethanol, 2,3-dihydroindole, isoindoline, 1,2,3, 4-tetrahydroisoquinoline, 1,2,3,4- tetrahydroquinoline, and the like; or can readily be prepared from commercially available starting materials using standard methods well known in the art, such as 4- hydroxyproline, 3-hydroxypiperidine, 4- hydroxypiperidine, 3-pyrroline, isonipecotic acid, 5- hydroxyindole, 5-hydroxyindole-3-acetic acid, 5,6- dimethoxyindole, isoquinoline, quinoline, 5- hydroxyisoquinoline, 8-hydroxy-5-nitroquinoline, 6,7- dimethoxy-1, 2, 3 ,4-tetrahydroiso
  • the amine is then coupled to the DL-, D-, or L- amino acid corresponding to the formula PNHCH(R 1 ) COOH, wherein P and R 1 are as defined above, followed by deprotection of the amine as described above and coupling the deprotected amine to the cyclic-amino acid of formula
  • n, R 10 , R 11 , R 12 , R 13 and R 14 are as defined above, such as pyrrolidin-1-ylacetic acid, piperidin-1- ylacetic acid and the like, to produce the antiviral compounds of the present invention.
  • the cyclic-amino acids are commercially available or are readily prepared from a protected carboxylic acid with a leaving group W (defined above) by reaction with the cyclic-amine as shown in Scheme III or alternatively, by reacting the cyclic-amine with the appropriate substituted ketone in the presence of cyanide anion followed by hydroylsis of the cyano group to the corresponding carboxylic acid as shown in Scheme IV, wherein n, P ,
  • R 14 are as defined above.
  • the amine is then coupled to the DL-, D-, or L- amino acid corresponding to the formula
  • n, R 1 , R 10 R 11 R 12 R 13 and R 14 are as defined above, which can be prepared in a similar fashion to the coupling methods described above from DL-, D-, or L- amino acid corresponding to the formula NH CH(R )C00P 3 , wherein P 3 and R 1 are as defined above.
  • the DL-, D-, or L-amino acid corresponding to the formula PNHCH(R 1 ) COOH or NH 2 CH(R 1 ) COOP 3 , wherein P, P 3 and R are as defined above, are commercially available (Sigma Chemical Co.), or readily prepared using standard methods well known in the art from readily available starting materials.
  • P is a benzyloxycarbonyl or t-butoxycarbonyl radical and P 3 is benzyl or tert-butyl radicals. Standard coupling procedures can be used to couple the amino acids and amines.
  • the carboxylic acid group is reacted to form an anhydride, mixed anhydride, acid halide, such as chloride or bromide, or active ester, such as esters of N-hydroxysuccinimide, HOBT and the like, using well known procedures and conditions.
  • acid halide such as chloride or bromide
  • active ester such as esters of N-hydroxysuccinimide, HOBT and the like
  • Appropriate solvent systems include tetrahydrofuran, ethylether, methyl- tert-butylether, methylene chloride, N,N- dimethylformamide and the like, including mixtures thereof .
  • the protected amino alcohol from the epoxide opening can be further protected at the newly introduced amino group with a protecting group P" which is not removed with the removal of the amino protecting groups P or P 1 and P 2 .
  • P P 1 , P, P 1 and P 2 .
  • n, P', R 1 , R 2 , R 3 , R 10 , R 11 , R 12 , R 13 and R 14 are as defined above.
  • the protecting group P' is then selectively removed and the resulting amine is reacted with the sulfonyl chloride RS0 2 C1, the ,sulfonyl bromide R S0 2 Br or the corresponding sulfonyl anhydride, preferably in the presence of an acid scavenger, to form the compounds of the present invention.
  • This selective deprotection and conversion to the sulfonamide can be accomplished at either the end of the synthesis or at any appropriate intermediate step if desired.
  • the benzylated phenylalanine phenylmethyl ester (0.302 mol) from the previous reaction was dissolved in toluene (750 mL) and cooled to -55°C.
  • a 1.5 M solution of DIBAL in toluene (443.9 mL, 0.666 mol) was added at a rate to maintain the temperature between -55 to -50°C (addition time -1 hr) .
  • the mixture was stirred for 20 minutes under a nitrogen atmosphere and then quenched at -55°C by the slow addition of methanol (37 ml) .
  • the cold solution was then poured into cold (5°C) 1.5 N HC1 solution (1.8 L) .
  • L-phenylalaninol (176.6 g, 1.168 mol) was added to a stirred solution of potassium carbonate (484.6 g, 3.506 mol) in 710 mL of water. The mixture was heated to 65°C under a nitrogen atmosphere. A solution of benzyl bromide (400 g, 2.339 mol) in 3A ethanol (305 mL) was added at a rate that maintained the temperature between 60-68°C. The biphasic solution was stirred at 65°C for 55 min and then allowed to cool to 10°C with vigorous stirring. The oily product solidified into small granules. The product was diluted with 2.0 L of tap water and stirred for 5 minutes to dissolve the inorganic by products.
  • the dichloromethane layer was separated from the aqueous phase and washed with water, brine, dried over magnesium sulfate, filtered and concentrated. The residue was diluted with ethyl acetate and hexane and then filtered to further remove the ammonium salt. The filtrate was concentrated to give ⁇ S- [bis (phenylmethyl) amino] benzenepropanaldehyde. The aldehyde was carried on to the next step without purification.
  • the mixture was cooled to 0 2C and an aqueous solution of 2.87 mL of 5% household bleach containing 0.735 g(8.75 mmoles) of sodium bicarbonate and 8.53 mL of water was added slowly over 25 minutes. The mixture was stirred at OSC for 60 minutes. Two more additions (1.44 mL each) of bleach was added followed by stirring for 10 minutes. The two phase mixture was allowed to separate. The aqueous layer was extracted twice with 20 mL of ethyl acetate.
  • the mixture was stirred for 10 minutes and the organic and aqueous layers were separated.
  • the aqueous phase was extracted with ethyl acetate (2x 500 mL) .
  • the ethyl acetate layer was combined with the tetrahydrofuran layer.
  • the combined solution was dried over magnesium sulfate (220g) , filtered and concentrated on a rotary evaporator at 65°C.
  • the brown oil residue was dried at 70°C in vacuo (0.8 bar) for 1 h to give 222.8g of crude material. (The crude product weight was >100%. Due to the relative instability of the product on silica gel, the crude product is usually used directly in the next step without purification) .
  • the diastereomeric ratio of the crude mixture was determined by proton NMR: (2S)/(2R): 86:14.
  • An analytical sample of each of the diastereomers was obtained by purification on silica-gel chromatography (3% ethyl acetate/hexane) and characterized as follows:
  • the product could be used in the next step without purification.
  • the desired diastereomer can be purified by recrystallization at a subsequent step.
  • the product could also be purified by chromatography.
  • the mixture was stirred for 10 minutes and the organic and aqueous layers were separated.
  • the aqueous phase was extracted with ethyl acetate (2x 500 mL) .
  • the ethyl acetate layer was combined with the tetrahydrofuran layer.
  • the combined solution was dried over magnesium sulfate (220g) , filtered and concentrated on a rotary evaporator at 65°C.
  • the brown oil residue was dried at 70°C in vacuo (0.8 bar) for 1 h to give 222.8g of crude material.
  • oxalic acid dihydrate (119g, 0.94 mole) was added to a 5000 mL round bottom flask fitted with a mechanical stirrer and a dropping funnel. Methanol (1000 ml) was added and the mixture stirred until dissolution was complete.
  • the brown oil was crystallized as follows: 0.2g of the oil was dissolved in the first solvent with heat to obtain a clear solution, the second solvent was added until the solution became cloudy, the mixture was heated again to clarity, seeded with about 99% diastereomerically pure product, cooled to room temperature and then stored in a refrigerator overnight. The crystals were filtered, washed with the second solvent and dried. The diastereomeric purity of the crystals was calculated from the HPLC peak areas. The results are shown in Table 1.
  • Methyl-tert-butylether (20.0 mL) was added to the oily residue and the mixture was stirred at room temperature for about 1 hour. Solid was isolated by filtration to afford 7.50g of crude salt. The crude salt was purified by recrystallization from ethyl acetate and heptane at room temperature to yield 4.13g (45.2% isomer recovery) of 95% diastereomerically pure L-tartaric acid salt (based on HPLC peak areas). Microanalysis: Calc. : C 67.76%, H 7.41%, N 4.94%; Found: C 70.06%, H 7.47%, N 5.07%.
  • the mixture was concentrated under reduced pressure and partitioned between 1 L of 2M ammonium hydroxide solution and 2 L of ether.
  • the ether layer was washed with 1 L of 1M ammonium hydroxide solution, twice with 500 mL water, 500 mL of brine and dried over magnesium sulfate for 1 hour.
  • the ether layer was filtered, concentrated under reduced pressure and the crude solid product was recrystallized from 110 mL of ethyl acetate and 1.3 L of hexane to give 115g (81% yield) of N-benzyl-L-phenylalaninol as a white solid.
  • N-benzyl-L-phenylalaninol (2.9g, 12 mmoles) was dissolved in 3 mL of triethylamine and 27 mL of methanol and 5.25g (24.1 mmoles) of di-tert-butyl dicarbonate was added. The mixture was warmed to 60°C for 35 minutes and concentrated under reduced pressure. The residue was dissolved in 150 mL of ethyl acetate and washed twice with 10 mL of cold (0-5°C) , dilute hydrochloric acid (pH 2.5 to 3), 15 mL of water, 10 mL of brine, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The crude product oil was purified by silica gel chromatography (ethyl acetate: hexane, 12:3 as eluting solvent) to give 3.98g (97% yield) of colorless oil.
  • the mixture was cooled to osc and an aqueous solution of 4.2 mL of 5% household bleach containing 0.23g (3.0 mL, 2.738 mmoles) of sodium bicarbonate was added slowly over 30 minutes.
  • the mixture was stirred at OSC for 10 minutes.
  • Three more additions (0.4 mL each) of bleach was added followed by stirring for 10 minutes after each addition to consume all the starting material.
  • the two phase mixture was allowed to separate.
  • the aqueous layer was extracted twice with 8 mL of toluene.
  • the combined organic layer was washed with 1.25 mL of a solution containing 0.075g of potassium iodide, sodium bisulfate(0.125 g) and water (1.1 mL) , 1.25 mL of 10% aqueous sodium thiosulfate solution, 1.25 mL of pH 7 phosphate buffer and 1.5 mL of brine solution.
  • the organic solution was dried over magnesium sulfate, filtered and concentrated under reduced pressure to give 0.32g (100% yield) of N-(t- Butoxycarbonyl) -N-benzyl-L-phenylalaninal.
  • This crude oil (>100% yield) is a mixture of the diastereomeric epoxides N, ⁇ S-bis (phenylmethyl) -N- (t- butoxycarbonyl) -2S-oxiranemethanamine and N, ⁇ S- bis (phenylmethyl) -N- (t-butoxycarbonyl) -2R- oxiranemethanamine.
  • the crude mixture is used directly in the next step without purification.
  • the organic layers were combined and washed with 100 mL water, 30 mL brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1.04g of a yellow crude oil.
  • the crude product was a mixture of the two diastereomers N, ⁇ S-bis (phenylmethyl) -N- (t- butoxycarbonyl) -2S-oxiranemethanamine and N, ⁇ S- bis (phenylmethyl) -N- (t-butoxycarbonyl) -2R- oxiranemethanamine.
  • Part A To a solution of 75. Og (0.226 mol) of N- benzyloxycarbonyl-L-phenylalanine chloromethyl ketone in a mixture of 807 mL of methanol and 807 mL of tetrahydrofuran at -2°C, was added 13.17g (0.348 mol, 1.54 equiv.) of solid sodium borohydride over one hundred minutes. The solvents were removed under reduced pressure at 40°C and the residue dissolved in ethyl acetate (approx. IL) . The solution was washed sequentially with 1M potassium hydrogen sulfate, saturated sodium bicarbonate and then saturated sodium chloride solutions.
  • Part B To a solution of 6.52g (0.116 mol, 1.2 equiv.) of potassium hydroxide in 968 L of absolute ethanol at room temperature, was added 32.3g (0.097 mol) of N-CBZ- 3 (S) -amino-l-chloro-4-phenyl-2 (S) -butanol. After stirring for fifteen minutes, the solvent was removed under reduced pressure and the solids dissolved in methylene chloride. After washing with water, drying over magnesium sulfate, filtering and stripping, one obtains 27.9g of a white solid.
  • Part C A solution of N-benzyloxycarboriyl 3(S)-amino- 1,2-(S) -epoxy-4-phenylbutane (l.OOg, 3.36 mmol) and isoamylamine (4.90g, 67.2 mmol, 20 equiv.) in 10 mL of isopropyl alcohol was heated to reflux for 1.5 hours. The solution was cooled to room temperature, concentrated in vacuo and then poured into 100 mL of stirring hexane whereupon the product crystallized from solution.
  • Part B Preparation of 2R-hydroxy-3- [[ (4-aminophenyl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propylamine
  • a solution of 3.0g (5.4 mmol) of carbamic acid, 2R- hydroxy-3-[ [ (4-nitrophenyl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester in 20 mL of ethyl acetate was hydrogenated over 1.5g of 10% palladium-on-carbon catalyst under 35 psig of hydrogen for 3.5 hours.
  • Part B Preparation of [2R-hydroxy-3-[ [ (3- aminophenyl) sulfonyl] (2-methylpropyl) amino]-lS- (phenylmethyl) propylamine
  • Part B Preparation of 5- (2 , 3-dihydrobenzofuranyl) sulfonyl chloride To a solution of 3.35g of anhydrous N,N- dimethylformamide at 0°C under nitrogen was added 6.18g of sulfuryl chloride, whereupon a solid formed. After stirring for 15 minutes, 4.69g of 2 , 3-dihydrobenzofuran was added, and the mixture heated at 100°C for 2 hours. The reaction was cooled, poured into ice water, extracted with methylene chloride, dried over magnesium sulfate, filtered and concentrated the crude material. This was recrystallized from ethyl acetate to afford 2.45g of 5- (2 , 3-dihydrobenzofuranyl) sulfonyl chloride.
  • reaction solution was then charged with a solution of 8.0g (19.1 mmol) of 2R-hydroxy-3-[ [ (2, 3- dihydrobenzofuran-5-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propylamine and 2.2g (22.0 mmol) of 4- methylmorpholine in 25 mL of anhydrous DMF and stirred for 15 hours.
  • the solvents were removed in vacuo and the residue was partitioned between 300 mL of ethyl acetate and 120 mL of 5% potassium hydrogen sulfate solution.
  • a Fischer-Porter bottle equipped with a magnetic stir bar was charged with 11.9g (17.9 mmol) of 2S- [ (carbobenzyloxy) amino] -N-[2R-hydroxy-3-[ (3- methylpropyl) (2 , 3-dihydrobenzofuran-5- * ylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S- meth lpentanamide and 75 mL of tetrahydrofuran (THF) .
  • the solution was hydrogenated in the presence of 5g of 10% palladium-on-carbon catalyst (50% water by weight) under 50 psig of hydrogen for 4 hours at room temperature.
  • the catalyst was removed by filtration, and the solvent removed in vacuo .
  • reaction solution was then charged with a solution of 8.8g (16.5 mmol) of 2S-amino-N-[2R-hydroxy- 3- [ (3-methylpropyl) (2,3-dihydro benzofuran-5- ylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S- methylpentanamide and 4.5g (44.1 mmol) of 4- methylmorpholine in 25 mL of anhydrous DMF and stirred for 16 hours. The solvents were removed in vacuo and the residue was partitioned between 300 mL of ethyl acetate and 120 mL of 5% potassium hydrogen sulfate solution.
  • Part B A solution of phenylmethyl [lS-[ [ [2R-hydroxy-3- [ (3-methyIbutyl) (phenylsulfonyl) -amino] -1S- (phenylmeth 1) propyl] amino] carbonyl] -2 , 2- dimethylpropyl]carbamate (200 mg, 0.31 mmol) in methanol (15 mL) was hydrogenated over 10% palladium on carbon for 2 hours. The reaction was filtered through diatomaceous earth and concentrated to an oil.
  • Part C The resulting free amine from part B (150 mg, 0.3 mmol) was combined with diisopropylethylamine
  • Part A To a solution of 2R-hydroxy-3-[N- (3- methyIbutyl) -N- (phenylsulfonyl) amino] -IS- (phenylmethyl) propylamine (2.79g, 7.1 mmol) in 27 mL of dioxane was added (2.3g, 7.1 mmol) of N-t-butylcarbonyl-L- isoleucine-N-hydroxysuccinamide ester, and the reaction was stirred under nitrogen atmosphere for 16 hours.
  • Part B (3.05g, 5.0 mmol) of the product from Part A was dissolved in 20 mL of 4N HC1 in dioxane and stirred under nitrogen atmosphere for 1.5 hours. The contents were concentrated in vacuo , and chased with diethyl ether. The crude hydrochloride salt was pumped on at 1 mm Hg until dry to yield 2.54g of product as its hydrochloride salt.
  • Part C (2.54g, 5.0 mmol) of amine hydrochloride was dissolved in 50 mL of tetrahydrofuran and to this was added (l.Olg, 10 mmol) of 4-methyl-morpholine, at which time a precipitate forms. To this suspension was added chloroacetic anhydride (0.865g, 5.0 mmol) and stirred for 40 minutes. The contents were concentrated in vacuo, and the residue partitioned in ethyl acetate (200 mL) and 5% KHS0 4 . The organic layer was washed with saturated sodium bicarbonate, and saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to yield the. crude product. Purification by silica gel chromatography using an eluant of 1:1 ethyl acetate: hexanes yielded 1.89g of pure chloroacetamide.
  • Part A To a cooled solution of N-t-Boc-L-isoleucine 2.31g (10 mmol) and 2.00g (13.11 mmol) of N- hydroxybenzotriazole in 17 mL of N,N-dimethylformamide was added 1.91g (10 mmol) of EDC and stirred at 0°C for one hour. To this was added a solution of 4.0g (10 mmol) of 2R-hydroxy-3-[ (2-methylpropyl) (4- methoxyphenylsulfonyl) amino] -IS-(phenylmethyl) propylamine in 6 mL of N,N-dimethylformamide and the solution stirred for 16 hours.
  • Part B 5.00g (8.0 mmol) of the product from Part A was dissolved in 20 mL of 4N HC1 in dioxane and stirred for 20 minutes. The precipitated product was stripped two times from diethyl ether to yield 2S- (amino) -N-[2R- hydroxy-3-[ (3-methylpropyl) (4-methoxyphenylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S- methylpentanamide'hydrochloride, which was used in Part C without further purification.
  • Part C The amine hydrochloride from part B was dissolved in 45 mL of methylene chloride and 3. Og N,N- diisopropylethylamme was added, followed by 1.22g (7.11 mmol) of chloroacetic anhydride. The solution was stirred at room temperature for 30 minutes. The contents were concentrated in vacuo and the residue was partitioned between ethyl acetate and water. The organic layer was washed with 5% citric acid, saturated sodium bicarbonate and brine.
  • Part D 4.8g (8.0 mmol) of 2S-[ (chloroacetyl) amino] -N- [2R-hydroxy-3-[ (3-methylpropyl) (4-methoxyphenylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S-methylpentanamide was dissolved in 15 mL of tetrahydofuran and to this was added water followed by 2.8 mL (40 mmol) of pyrrolidine and the reaction stirred for 1.5 hours. The solvents were removed in vacuo and the residue was dissolved in ethyl acetate.
  • Method 1 Carbamic acid, 2R-hydroxy-3-[ [ (2- aminobenzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl-, phenylmethyl ester (0.25g, 0.429 mmol) was added to a solution of isoamylnitrite (0.116 mL, 0.858 mmol) in dioxane (5 mL) and the mixture was heated at 85°C. After the cessation of evolution of nitrogen, the reaction mixture was concentrated and the residue was purified by chromatography (hexane: ethyl acetate 5:3) to afford 0.130g (53%) of the desired product as a solid.
  • reaction mixture was diluted with ethyl acetate (100 mL) , washed with citric acid (5%, 100 mL) , sodium bicarbonate (saturated, 100 mL) and brine (100 mL) , dried (MgS04) and concentrated in vacuo .
  • citric acid 5%, 100 mL
  • sodium bicarbonate saturated, 100 mL
  • brine 100 mL
  • the residue was chromatographed (silica gel, ethyl acetate: hexane 1:1) to afford 0.340g (23%) of desired product.
  • the carbamic acid, 2R-hydroxy-3-[ (3-aminophenylsulfonyl) (2-methylpropyl) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester 0.36g (0.685 mmol) was added to a well mixed powder of anhydrous copper sulfate (1.44 g) and potassium thiocyanate (1.80 g) followed by dry methanol (10 mL) and the resulting black-brown suspension was heated at reflux for 2 hrs. The reaction mixture was filtered and the filtrate was diluted with water (5 mL) and heated at reflux. Ethanol was added to the reaction mixture, cooled and filtered.
  • Part B 3 (S) -[N- (benzyloxycarbonyl) amino]-l, 2 (S) -epoxy- 4-phenylbutane
  • Part D phenylmethyl [2 (R) -hydroxy-3-[N- (2- methylpropyl) -N- (4-methoxyphenylsulfonyl) amino] -1S- (phenylmethyl) propyl] carbamate
  • Method 1 To a solution of 4.25g of anhydrous N,N- dimethylformamide at 0°C under nitrogen was added 7.84g of sulfuryl chloride, whereupon a solid formed. After stirring for 15 minutes, 6.45g of 1, 3-benzodioxole was added, and the mixture heated at 100°C for 2 hours. The reaction was cooled, poured into ice water, extracted with methylene chloride, dried over magnesium sulfate, filtered and concentrated to give 7.32g of crude material as a black oil. This was chromatographed on silica gel using 20% methylene chloride/hexane to afford 1.9g of (l,3-benzodioxol-5-yl) sulfonyl chloride.
  • the mixture was heated to 67 °C for 5 hours followed by cooling to 16 °C with an ice bath.
  • the reaction was quenched with water (51) at a rate which kept the temperature below 20°C. After the addition of water was complete, the mixture was stirred for 10 minutes.
  • the layers were separated and the organic layer was washed again twice with water (51) .
  • the organic layer was dried with magnesium sulfate (500g) and filtered to remove the drying agent. The solvent was removed under vacuum at 50°C. The resulting warm liquid was allowed to cool at which time a solid began to form. After one hour, the solid was washed with hexane (400 L) , filtered and dried to provide the desired sulfonyl chloride (2823g) .
  • the hexane wash was concentrated and the resulting solid washed with 400 mL hexane to provide additional sulfonyl chloride (464g) .
  • the total yield was 3287g (95.5% based upon 1,3- benzodioxole) .
  • Method 3 Dichloroethane (DCE) , 2 liters, was added to a 30 L reactor fitted with condenser (0°C) , two agitator mechanical stirrer, internal cooling coils heating mantel and nitrogen atmosphere/purge system. Sulfur trioxide «DMF complex (4856g, 31.7 moles) was then added over a 15 minute period. No temperature rise was observed. The reaction mixture was heated to 75°C. The material was completely dissolved in two hours. NMR analysis indicated that the reaction was complete in less than 5 hours. The reaction was allowed to cool overnight for convenience. The reaction mass was cooled to 15°C. A caustic scrubber (a 4L Erlenmeyer flask containing 2 L of 25% NaOH with subsurface addition) was attached to the top of the condenser. (The scrubber was changed when NaCl precipitation was significant) .
  • DCE dichloroethane
  • Oxalyl chloride (4041g, 31.84 moles) was added over a 1.5 hour period while maintaining the temperature between 11° and 15°C.
  • the reaction mixture was allowed to come to room temperature over 30 minutes at which time gas evolution had ceased. NMR analysis after an additional hour at room temperature indicated that the reaction was complete.
  • the reaction mass was then cooled to 15°C and water (2 L) was added very slowly while keeping the temperature below 25°C.
  • the reaction mixture was stirred for 10 minutes. The stirring was stopped and the layers were allowed to separate. The water layer was removed and the layers allowed to separate.
  • the water layer was removed and the organic" layer was washed twice with water (8 L) .
  • the organic layer was then dried over magnesium sulphate (1 Kg) which clarified the solution.
  • the solution was filtered through a sintered glass funnel in two batches and the residue from each was washed with DCE (1 L) .
  • the solvent was removed at 50°C in vacuo (ca. 100 mm of Hg to remove the majority of the solvent then 2 mm of Hg) .
  • the material solidified as it cooled to room temperature to yield the desired sulfonyl chloride (5337g, 91.65% yield) which was used in the subsequent step without further purification.
  • Method 1 To a 5000 mL, 3-necked flask fitted with a mechanical stirrer was added N-[3 (S) -[N,N- bis (phenylmethyl) amino] -2 (R) -hydroxy-4-phenylbuty1] -N- isobutylamine»oxalic acid salt (354.7g, 0.7 mole) and 1,4-dioxane (2000 mL) . A solution of potassium carbonate (241.9g, 1.75 moles) in water (250 mL) was then added.
  • the bottle was placed in the hydrogenator and purged 5 times with nitrogen and 5 times with hydrogen. The reaction was allowed to proceed at 35 °C with 63 PSI hydrogen pressure for 18 hours. Additional catalyst (125 mg) was added and, after purging, the hydrogenation continued for and additional 20 hours.
  • the mixture was filtered through celite which was washed with methanol (2 X 10 mL) . Approximately one third of the methanol was removed under reduced pressure. The remaining methanol was removed by aziotropic distillation with toluene at 80 torr. Toluene was added in 15, 10, 10 and 10 mL portions. The product crystallized from the mixture and was filtered and washed twice with 10 mL portions of toluene. The solid was dried at room temperature at 1 torr for 6 hours to yield the amine salt (4.5g, 84%). HPLC/MS (electrospray) was consistent with the desired product (m/z 421 [M+H] + ) .
  • Part B To the THF solution of crude 1-[N-[(1,3- benzodioxol-5-y1) sulfonyl] -N- (2-methylpropyl) amino] - 3 (S) -[bis (phenylmethyl) amino] -4-phenyl-2 (R) -butanol was added water (500 mL) followed by methane sulfonic acid (531g, 5.5 moles). The solution was stirred to insure complete mixing and added to a 5 gallon autoclave. Pearl an's catalyst (200g of 20% Pd(0H) 2 on C/ 50% water) was added to the autoclave with the aid of THF (500 mL) .
  • the reactor was purged four times with nitrogen and four times with hydrogen.
  • the reactor was charged with 60 psig of hydrogen and stirring at 450 rpm started. After 16 hours, HPLC analysis indicated that a small amount of the mono-benzyl intermediate was still present.
  • Additional catalyst 50g was added and the reaction was allowed to run overnight.
  • the solution was then filtered through celite (500g) to remove the catalyst and concentrated under vacuum in five portions. To each portion, toluene (500 mL) was added and removed under vacuum to azeotropically removed residual water. The resulting solid was divided into three portions and each washed with methyl t-butyl ether (2 L) and filtered. The residual solvent was removed at room temperature in a vacuum oven at less than 1 torr to yield the 2714g of the expected salt.
  • the product can be further purified by the following procedure.
  • a total of 500 mL of methanol and 17Og of material from above was heated to reflux until it all dissolved.
  • the solution was cooled, 200 mL of isopropanol added and then 1000-1300 mL of hexane, whereupon a white solid precipitated. After cooling to 0°C, this precipitate was collected and washed with hexane to afford 123g of the desired material.
  • the original material which was a 95:5 mixture of alcohol diastereomers was greater than 99:1 of the desired diastereomer.
  • Part B Preparation of 2R-hydroxy-3-[ [ (1, 3-benzodioxol- 5-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propylamine
  • a mixture of sulfanilamide (86g, 0.5 mole), ammonium thiocyanate (76. Og, 0.5 mole) and dilute hydrochloric acid (1.5 N, 1 L) was mechanically stirred and heated at reflux for 2 hr. About 200 mL of water was distilled off and concentration of the reaction mixture afforded a solid. The solid was filtered and was washed with cold water and air dried to afford 67.5g (59%) of the desired product as a white powder.
  • Part B Preparation of N-[2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-y1) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-amino-3 , 3-dimethylbutanamide
  • Part D Preparation of N- [2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl] -2S-[ [ (pyrrolidin-l- yl) acetyl]amino] -3, 3-dimethylbutanamide
  • N-t-Boc-L-isoleucine 2.02g (8.74 mmol) and 2.00g (13.11 mmol) of N-hydroxybenzotriazole in 17 mL of N,N-dimeth Iformamide was added 1.84g (9.61 mmol) of EDC and stirred at 0°C for one hour.
  • Part D Preparation of N- [2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl-amino] -1S- (phenylmethy1) propyl] -2S- [ [ (pyrrolidino) acetyl] amino] - 3S-methylpentanamide
  • Part B Preparation of N-[2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl] -2S-amino-3S-methylpentanamide
  • a Fischer-Porter bottle equipped with a magnetic stir bar was charged with 10.2g (16.4 mmol) of the product from Part A and 75 mL of tetrahydrofuran (THF) .
  • the solution was hydrogenated in the presence of 4g of 10% palladium-on-carbon catalyst (50% water by weight) under 50 psig of hydrogen for 3 hours at room temperature.
  • the catalyst was removed by filtration, and the solvents removed under reduced pressure.
  • N-t-Boc-S-methyl-(L) -cysteine (2.80 g, 11.9 mmol)
  • 1- Hydroxybenzotriazole hydrate (1.92 g, 12.5 mmol)
  • 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (2.27 g, 11.9 mmol) were mixed in N,N- dimethyIformamide (30.0 mL) at 0°C for 10 min.
  • N- Methylmorpholine (3.03 g, 33.0 mmol) was added and the solution stirred an additional 10 min at 0°C.
  • Part B Preparation of N-[ [2R-hydroxy-3-[ (1,3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S- [ [ (1, 1-dimethylethoxy) carbonyl] amino] -3- (methylsulfonyl) propaneamide
  • Part D Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S- [ (chloroacetyl) amino] -3- (methylsulfonyl) propaneamide
  • Part E Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -3- (methylsulfonyl) propaneamide
  • N-t-boc-S-methyl-L-penicillamine dicyclohexylamine salt (4.00g, 9.00 mmol), 1-Hydroxybenzotriazole hydrate (1.69 g, 11.00 mmol), and l- (3-dimethylaminopropyl) -3- ethylcarbodiimide hydrochloride (1.71 g, 9.00 mmol) were mixed in dimethylformamide (60.0 mL) at room temperature.
  • the heterogeneous mixture was stirred for 1 hour and 2R-hydroxy-3-[ [ (1, 3-benzodioxol-5- yl) sulfonyl] (2-methylpropyl) amino] -1S-
  • Part B Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-[ [ (1, l-dimethylethoxy) carbonyl] amino] -3-methy1-3- (methylsulfonyl) butaneamide
  • Part E Preparation of N-[ [2R-hydroxy-3-[ (1,3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl]-2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -3-methy1-3-(methylsulfonyl) butaneamide
  • Part B Preparation of N- [2R-hydroxy-3- [ [ (1, 1- dimethylethoxy) carbonyl] (2-methylpropyl) amino]-lS- (phenylmethyl) propyl] -2S- [ (phenylmethoxycarbonyl) amino] - 3 , 3-dimethylbutanamide
  • N-[ (1, l-dimethylethoxy1) carbonyl] -N-[2-methylpropyl] -3S- [N 1 - (phenyImethoxycarbony1) amino] -2R-hydroxy-4- phenylbutylamine in ethanol was hydrogenated at 45 psig of hydrogen in the presence of 5% pd(C) catalyst to yield N-[ (1, l-dimethylethoxy1) carbonyl]-N-[2- methylpropyl] -3S- [N 1 - (phenylmethoxycarbonyl) amino] -2R- hydroxy-4-phenylbutylamine.
  • reaction mixture was stirred for 18 hours.
  • the reaction mixture was concentrated and the residue was dissolved in ethyl acetate (500 mL) , washed with sodium bicarbonate (saturated, 200 mL) and brine (200 mL) , dried (MgS04) and concentrated to afford 11.0 g (quantitative) of the desired product.
  • Part B Preparation of N-[2R-hydroxy-3- [ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-1- yl) acetyl] amino] -3 , 3-dimethylbutanamide»dihydrochloride
  • reaction mixture was concentrated and the residue was dissolved in dichloromethane (200 mL) and was washed with sodium hydroxide (IN, 100 mL) , citric acid (5%, 100 mL) dried (Mg S04) and concentrated to afford a residue which was chromatographed (ethyl acetate:hexane 3:1) to afford 1.8 g (65%) of the desired product as a solid.
  • the compounds of the present invention are effective HIV protease inhibitors. Utilizing an enzyme assay as described below, the compounds set forth in the examples herein disclosed inhibited the HIV enzyme.
  • the preferred compounds of the present invention and their calculated IC 50 (inhibiting concentration 50%, i.e., the concentration at which the inhibitor compound reduces enzyme activity by 50%) values are shown in Tables 20 and 21.
  • the enzyme method is described below.
  • the substrates is 2-Ila-Nle-Pha (p-N0 2 ) ) Gln-ArgJH 2 .
  • the positive control is MVT-101 (Miller, M. et al, Science, 246, 1149 (1989)]
  • the assay conditions are as follows: Assay buffer: 20 M sodium phosphate, pH 6.4 20% glycerol 1 mM EDTA
  • the final concentration of DMSO is 14% and the final concentration of glycerol is 18%.
  • the test compound is dissolved in DMSO and diluted in DMSO to lOx the test concentration; lO ⁇ l of the enzyme preparation is added, the materials mixed and then the mixture is incubated at ambient temperature for 15 minutes.
  • the enzyme reaction is initiated by the addition of 40 ⁇ l of substrate.
  • the increase in fluorescence is monitored at 4 time points (0, 8, 16 and 24 minutes) at ambient temperature. Each assay is carried out in duplicate wells.
  • the effectiveness of various compounds were determined in the above-described enzyme assay and in a CEM cell assay.
  • the HIV inhibition assay method of acutely infected cells is an automated tetrazolium based colorimetric assay essentially that reported by Pauwles et al, J. Virol. Methods, 20, 309-321 (1988) .
  • Assays were performed in 96-well tissue culture plates. CEM cells, a CD4 + cell line, were grown in RPMI-1640 medium (Gibco) supplemented with a 10% fetal calf serum and were then treated with polybrene (2 ⁇ g/ml) . An 80 ⁇ l volume of medium containing 1 x 10 cells was dispensed into each well of the tissue culture plate.
  • tissue culture medium or medium without test compound as a control
  • TCID 50 the dose of virus that infects 50% of cells in tissue culture
  • 20 ⁇ L volume of the virus sample was added to wells containing test compound and to wells containing only medium (infected control cells) .
  • Several wells received culture medium without virus (uninfected control cells) .
  • the intrinsic toxicity of the test compound was determined by adding medium without virus to several wells containing test compound.
  • the tissue culture plates contained the following experiments:
  • test compounds were 1, 10, 100 and 500 ⁇ g/ l. Either azidothymidine (AZT) or dideoxyinosine (ddl) was included as a positive drug control. Test compounds were dissolved in DMSO and diluted into tissue culture medium so that the final DMSO concentration did not exceed 1.5% in any case. DMSO was added to all control wells at an appropriate concentration.
  • test compounds could be added on days 0, 2 and 5 if desired.
  • day 7 post-infection, the cells in each well were resuspended and a lOO ⁇ l sample of each cell suspension was removed for assay.
  • a 20 ⁇ L volume of a 5 mg/ml solution of 3-(4,5-dimethylthiazol-2-yl) -2,5- diphenyltetrazolium bromide (MTT) was added to each lOO ⁇ L cell suspension, and the cells were incubated for 4 hours at 27°C in a 5% C0 2 environment.
  • MTT is metabolically reduced by living cells resulting in the production in the cell of a colored formazan product.
  • To each sample was added lOO ⁇ l of 10% sodium dodecylsulfate in 4.01 N HG1 to lyse the cells, and samples were incubated overnight. The absorbance at 590 n was determined for each sample using a Molecular Devices icroplate reader. Absorbance values for each set of wells is compared to assess viral control infection, uninfected control cell response as well as test compound by cytotoxicity and antiviral efficacy.
  • the compounds of the present invention are effective antiviral compounds and, in particular, are effective retroviral inhibitors as shown above.
  • the subject compounds are effective HIV protease inhibitors. It is contemplated that the subject compounds will also inhibit other retroviruses such as other lentiviruses in particular other strains of HIV, e.g. HIV-2 , human T-cell leukemia virus, respiratory syncitial virus, simia immunodeficiency virus, feline leukemia virus, feline immuno-deficiency virus, hepadnavirus, cytomegalovirus and picornavirus.
  • the subject compounds are effective in the treatment, proplylaxis of retroviral infections and/or the prevention of the spread of retroviral infections.
  • the subject compounds are also effective in preventing the growth of retroviruses in a solution.
  • Both human and animal cell cultures such as T- lymphocyte cultures, are utilized for a variety of well known purposes, such as research and diagnostic procedures including calibrators and controls.
  • the subject compounds Prior to and during the growth and storage of a cell culture, the subject compounds may be added to the cell culture medium at an effective concentration to prevent the unexpected or undesired replication of a retrovirus that may inadvertently, unknowingly or knowingly be present in the cell culture.
  • the virus may be present originally in the cell culture, for example HIV is known to be present in human T-lymphocytes long before it is detectable in blood, or through exposure to the virus.
  • Compounds of the present invention can possess one or more asymmetric carbon atoms and are thus capable of existing in the form of optical isomers as well as in the form of racemic or nonracemic mixtures thereof.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example by formation of diastereoisomeric salts by treatment with an optically active acid or base.
  • Examples of appropriate acids are tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric and camphorsulfonic acid and then separation of the mixture of diastereoisomers by crystallization followed by liberation of the optically active bases from these salts.
  • a different process for separation of optical isomers involves the use of a chiral chromatography column optimally chosen to maximize the separation of the enantiomers.
  • Still another available method involves synthesis of covalent diastereoisomeric molecules by reacting compounds of Formula I with an optically pure acid in an activated form or an optically pure isocyanate.
  • the synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound.
  • the optically active compounds of Formula I can likewise be obtained by utilizing optically active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt.
  • the compounds of the present invention can be used in the form of salts derived from inorganic or organic acids.
  • These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobro ide, hydroiodide, 2-hydroxy-ethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2- naphthalenesulfonate, oxalate, palmoate, pectinate, persulfate,
  • the basic nitrogen- containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl"bromides, and others. Water or oil-soluble or dispersible products are thereby obtained.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates
  • acids which may be employed to form pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, sulphuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, succinic acid and citric acid.
  • organic acids such as oxalic acid, maleic acid, succinic acid and citric acid.
  • Other examples include salts with alkali metals or alkaline earth metals, such as sodium, potassium, calcium or magnesium or with organic bases.
  • Total daily dose administered to a host in single or divided doses may be in amounts, for example, from 0.001 to 10 mg/kg body weight daily and more usually 0.01 to 1 mg. Dosage unit compositions may contain such amounts of submultiples thereof to make up the daily dose.
  • 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.
  • the dosage regimen for treating a disease condition with the compounds and/or compositions of this invention is selected in accordance with a variety of factors, including the type, age, weight, sex, diet and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetic and toxicology profiles ⁇ of the particular compound employed, whether a drug delivery system is utilized and whether the compound is administered as part of a drug combination.
  • the dosage regimen actually employed may vary widely and therefore may deviate from the preferred dosage regimen set forth above.
  • the compounds of the present invention may be administered orally, parenterally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. Topical administration may also involve the use of transdermal administration such as transdermal patches or iontophoresis devices.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection, or infusion techniques.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol .
  • a nontoxic parenterally acceptable diluent or solvent for example, as a solution in 1, 3-butanediol .
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic 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 find use in the preparation of injectables.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable nonirritating excipient such as cocoa butter and polyethylene glycols which are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • a suitable nonirritating excipient such as cocoa butter and polyethylene glycols which are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules.
  • the active compound may be admixed with at least one inert diluent such as sucrose lactose or starch.
  • Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g. , lubricating agents such as magnesium stearate.
  • the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring, and perfuming agents. While the compounds of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more immunomodulator ⁇ , antiviral agents or other antiinfective agents.
  • the compounds of the invention can be administered in combination with AZT, DDI, DDC or with glucosidase inhibitors, such as N-butyl-1-deoxynojirimycin or prodrugs thereof, for the prophylaxis and/or treatment of AIDS.
  • the therapeutic agents can be formulated as separate compositions which are given at the same time or different times, or the therapeutic agents can be given as a single composition.

Abstract

Process for preparing a benzo fused heterocyclo sulfonyl halide intermediate, comprising reacting a benzo fused heterocyclic compound with a -SO3- complex in the presence of a solvent and then adding oxalyl halide.

Description

PROCESS FOR THE PREPARAΗON OF A BENZO FUSED HETEROCYCLO SULFONYL HALIDE INTERMEDIATE
BACKGROUND OF THE INVENTION
The present invention relates to retroviral 5 protease inhibitors and, more particularly, relates to novel compounds, composition and method for inhibiting retroviral proteases, such as human immunodeficiency virus (HIV) protease. This invention, in particular, relates to a ino acid hydroxyethylamine sulfonamide 10 protease inhibitor compounds, composition and method for inhibiting retroviral proteases, prophylactically preventing retroviral infection or the spread of a retrovirus, and treatment of a retroviral infection, e.g., an HIV infection. The subject invention also 15 relates to processes for making such compounds as well as to intermediates useful in such processes.
During the replication cycle of retroviruses, gag and gag-pol gene transcription products are translated as proteins. These proteins are subsequently processed
20 by a virally encoded protease (or proteinase) to yield viral enzymes and structural proteins of the virus core. Most commonly, the gag precursor proteins are processed into the core proteins and the pol precursor proteins are processed into the viral enzymes, e.g.,
25 reverse transcriptase and retroviral protease. It has been shown that correct processing of the precursor proteins by the retroviral protease is necessary for assembly of infectious virons. For example, it has been shown that frameshift mutations in the protease region of the pol gene of HIV prevents processing of the gag precursor protein. It has also been shown through site-directed mutagenesis of an aspartic acid residue in the HIV protease active site that processing of the gag precursor protein is prevented. Thus, attempts have been made to inhibit viral replication by inhibiting the action of retroviral proteases.
Retroviral protease inhibition typically involves a transition-state mimetic whereby the retroviral protease is exposed to a mimetic compound which binds (typically in a reversible manner) to the enzyme in competition with the gag and gag-pol proteins to thereby inhibit specific processing of structural proteins and the release of retroviral protease itself. In this manner, retroviral replication proteases can be effectively inhibited.
Several classes of compounds have been proposed, particularly for inhibition of proteases, such as for inhibition of HIV protease. Such compounds include hydroxyethylamine isosteres and reduced amide isosteres. See, for example, EP 0346847; EP 0342541; Roberts et al, "Rational Design of Peptide-Based Proteinase Inhibitors, "Science, 248, 358 (1990); and Erickson et al, "Design Activity, and 2. δA Crystal Structure of a C2 Symmetric
Inhibitor Complexed to HIV-1 Protease," Science, 249, 527 (1990). US 5,157,041, WO 94/04491, WO 94/04492, WO 94/04493, WO 94/05639, WO 92/08701 and US Patent Application Serial No. 08/294,468, filed August 23, 1994, (each of which is incorporated herein by reference in its entirety) for example describe hydroxyethylamine, hydroxyethylurea or hydroxyethyl sulfonamide isostere containing retroviral protease inhibitors.
Several classes of compounds are known to be useful as inhibitors of the proteolytic enzyme renin. See, for example, U.S. No. 4,599,198; U.K. 2,184,730; G.B. 2,209,752; EP 0264795; G.B. 2,200,115 and U.S. SIR H725. Of these, G.B. 2,200,115, GB 2,209,752, EP 0264795, U.S. SIR H725 and U.S. 4,599,198 disclose urea-containing hydroxyethylamine renin inhibitors. EP 468641 discloses renin inhibitors and intermediates for the preparation of the inhibitors, which include sulfonamide-containing hydroxyethylamine compounds, such as 3- (t-butoxycarbonyl) amino-cyclohexyl-1-
(phenylsulfonyl)amino-2 (5)-butanol. G.B. 2,200,115 also discloses sulfamoyl-containing hydroxyethylamine renin inhibitors, and EP 0264795 discloses certain sulfonamide-containing hydroxyethylamine renin inhibitors. However, it is known that, although renin and HIV proteases are both classified as aspartyl proteases, compounds which are effective renin inhibitors generally are not predictive for effective HIV protease inhibition.
BRIEF DESCRIPTION OF THE INVENTION
The present invention relates to selected retroviral protease inhibitor compounds, analogs and pharmaceutically acceptable salts, esters and prodrugs thereof. The subject compounds are characterized as amino acid hydroxyethylamine sulfonamide inhibitor compounds. The invention compounds advantageously inhibit retroviral proteases, such as human immunodeficiency virus (HIV) protease. Therefore, this invention also encompasses pharmaceutical compositions, methods for inhibiting retroviral proteases and methods for treatment or prophylaxis of a retroviral infection, such as an HIV infection. The subject invention also relates to processes for making such compounds as well as to intermediates useful in such processes.
DETAILED DESCRIPTION OF THE INVENTION
In accordance with the present invention, there is provided a retroviral protease inhibiting compound of the formula:
Figure imgf000006_0001
or a pharmaceutically acceptable salt, prodrug or ester thereof, wherein n represents 1 or 2;
R1 represents alkyl, alkenyl, alkynyl, hydroxyalkyl, alkoxyalkyl, cyanoalkyl, imidazolylmethyl, -CH2CONH2, -CH2CH2CONH2, -CH2S(0)2NH2 -CH2SCH3, -CH2S(0)CH3, -CH2S(0)2CH3, -C(CH3)2SCH3, -C (CH3) 2S (0) CH3 or -(CH3)2S(0)2CH3 radicals; preferably, R1 represents alkyl of 1-5 carbon atoms, alkenyl of 2-5 carbon atoms, alkynyl of 2-5 carbon atoms, hydroxyalkyl of 1-3 carbon atoms, alkoxyalkyl of 1-3 alkyl and 1-3 alkoxy carbon atoms, cyanoalkyl of 1-3 alkyl carbon atoms, imidazolylmethyl, -CH2C0NH2, -CH2CH2CONH2 , -CH2S (0) 2NH2 , -CH2SCH3, -CH2S(0)CH3, -CH S (0) 2CH3 , -C (CH3) 2SCH3 , -C(CH3)2S(0)CH3 or -C (CH3) 2S (0) 2CH3 radicals; more preferably, R1 represents alkyl of 1-4 carbon atoms, alkenyl of 2-3 carbon atoms, alkynyl of 3-4 carbon atoms, cyanomethyl, imidazolylmethyl, -CH2CONH2, -CH2CH2CONH2 , -CH2S(0)2NH2, -CH2SCH3, -CH2S(0)CH3, -CH2S(0)2CH3, -C(CH3)2SCH3, -C (CH3) 2S (0) CH3 or -C(CH3)2S(0)2CH3 radicals; and most preferably, R1 represents sec-butyl, tert-butyl, iso-propyl, 3- propynyl or -C(CH3) S (0) 2CH3 radicals;
R2 represents alkyl, aralkyl, alkylthioalkyl, arylthioalkyl or cycloalkylalkyl radicals; preferably,
R represents radicals of alkyl of 1-5 carbon atoms, aralkyl of 1-3 alkyl carbon atoms, alkylthioalkyl of 1-
3 alkyl carbon atoms, arylthioalkyl of 1-3 alkyl carbon atoms or cycloalkylalkyl of 1-3 alkyl carbon atoms and , 3-6 ring member carbon atoms; more preferably, R represents radicals of alkyl of 3-5 carbon atoms, arylmethyl, alkylthioalkyl of 1-3 alkyl carbon atoms, arylthiomethyl or cycloalkylmethyl of 5-6 ring member carbon atoms radicals; even more preferably, R represents isobutyl, n-butyl, CH3SCH2CH2-, benzyl, phenylthiomethyl, (2-naphthylthio)methyl, 4-methoxy phenylmethyl, 4-hydroxyphenylmethyl, 4- fluorophenylmethyl or cyclohexylmethyl radicals; even more preferably, R2 represents benzyl, 4- fluorophenylmethyl or cyclohexylmethyl radicals; most preferably, R2 represents benzyl;
R3 represents alkyl, cycloalkyl or cycloalkylalkyl radicals; preferably, R3 represents radicals of alkyl radical of 1-5 carbon atoms, cycloalkyl of 5-8 ring members or cycloalkylmethyl radical of 3-6 ring members; more preferably, R3 represents propyl, isoamyl, isobutyl, butyl, cyclopentylmethyl, cyclohexylmethyl, cyclohexyl or cycloheptyl radicals; more preferably R3 represents isobutyl or cyclopentylmethyl radicals; R4 represents aryl, heteroaryl or heterocyclo radicals; preferably, R represents aryl, benzo fused 5 to 6 ring member heteroaryl or benzo fused 5 to 6 ring member heterocyclo radicals; or
R4 represents a radical of the formula
Figure imgf000008_0001
wherein A and B each independently represent 0, S, SO or S0 ; preferably, A and B each represent 0;
R6 represents hydrogen deuterium, alkyl or halogen radicals; preferably, R6 represents hydrogen deuterium, alkyl of 1-5 carbon atoms, fluoro or chloro radicals; more preferably R6 represents hydrogen deuterium, methyl, ethyl, propyl, isopropyl or fluoro radicals;
R7 represents hydrogen, deuterium, alkyl or halogen radicals; preferably, R7 represents hydrogen, deuterium, alkyl of 1-3 carbon atoms, fluoro or chloro radicals; more preferably, R7 represents hydrogen, deuterium, methyl or fluoro radicals; or R6 and R7 each independently represent fluoro or chloro radicals; and preferably, R6 and R7 each represent a fluoro radical; or
R represents a radical of the formula
Figure imgf000008_0002
wherein Z represents 0, S or NH; and R9 represents a radical of formula
Figure imgf000009_0001
wherein Y represents 0, S or NH; X represents a bond, 0 or NR21;
R20 represents hydrogen, alkyl, alkenyl, alkynyl, aralkyl, heteroaralkyl, heterocycloalkyl, a inoalkyl, N- mono-substituted or N,N-disubstituted aminoalkyl wherein said substituents are alkyl or aralkyl radicals, carboxyalkyl, alkoxycarbonylalkyl, cyanoalkyl or hydroxyalkyl radicals; preferably, R20 represents hydrogen, alkyl of 1 to 5 carbon atoms, alkenyl of 2 to 5 carbon atoms, alkynyl of 2 to 5 carbon atoms, aralkyl of 1 to 5 alkyl carbon atoms, heteroaralkyl of 5 to 6 ring members and 1 to 5 alkyl carbon atoms, heterocycloalkyl of 5 to 6 ring members and 1 to 5 alkyl carbon atoms, aminoalkyl of 2 to 5 carbon atoms, N-mono- substituted or N,N-disubstituted aminoalkyl of 2 to 5 alkyl carbon atoms wherein said substituents are radicals of alkyl of 1 to 3 carbon atoms, aralkyl of 1 to 3 alkyl carbon atoms radicals, carboxyalkyl of 1 to 5 carbon atoms, alkoxycarbonylalkyl of 1 to 5 alkyl carbon atoms, cyanoalkyl of 1 to 5 carbon atoms or hydroxyalkyl of 2 to 5 carbon atoms; more preferably, R20 represents hydrogen, alkyl of 1 to 5 carbon atoms, phenylalkyl of 1 to 3 alkyl carbon atoms, heterocycloalkyl of 5 to 6 ring members and 1 to 3 alkyl carbon atoms, or N-mono- substituted or N,N-disubstituted aminoalkyl of 2 to 3 carbon atoms wherein said substituents are alkyl radicals of 1 to 3 carbon atoms; and most preferably, R20 represents hydrogen, methyl, ethyl, propyl, isopropyl, isobutyl, benzyl, 2-(l-pyrrolidinyl) ethyl, 2- (1-piperidinyl) ethyl, 2-(l-piperazinyl) ethyl, 2- (4- methylpiperazin-1-yl) ethyl, 2- (1-morpholinyl) ethyl, 2- (1-thiamorpholinyl) ethyl or 2- (N,N-dimethylamino) ethyl radicals;
R21 represents hydrogen or alkyl radicals; preferably, R21 represents hydrogen radical or alkyl radical of 1 to 3 carbon atoms; more preferably, R21 represents hydrogen or methyl radicals; and most preferably, R21 represents a hydrogen radical; or
the radical of formula -NR20R21 represents a heterocyclo radical; preferably, the radical of formula -NR20R21 represents a 5 to 6 ring member heterocyclo radical; more preferably, the radical of formula -NR20R21 represents pyrrolidinyl, piperidinyl, piperazinyl, 4- methylpiperazinyl, 4-benzylpiperazinyl, morpholinyl or thiamorpholinyl radicals; and
R22 represents alkyl or R20R21N-alkyl radicals; preferably, R22 represents alkyl or R20R21N-alkyl radicals wherein alkyl is 1 to 3 carbon atoms; and more preferably, R22 represents alkyl radical of 1 to 3 carbon atoms; and
preferably R4 represents phenyl, 2-naphthyl, 4- methoxyphenyl , 4-hydroxyphenyl, 3 ,4-dimethoxyphenyl, 3- aminophenyl, 4-aminophenyl, benzothiazol-5-yl, benzothiazol-6-yl, 2-amino-benzothiazol-5-yl, 2- (methoxycarbonylamino)benzothiazol-5-yl, 2-amino- benzothiazol-6-yl, 2- (methoxycarbonylamino) benzothiazol-6-yl, 5-benzoxazolyl, 6-benzoxazolyl, 6- benzopyranyl, 3 , 4-dihydrobenzopyran-6-yl, 7- benzopyranyl, 3 , 4-dihydrobenzopyran-7-yl, 2,3- dihydrobenzofuran-5-yl, benzofuran-5-yl, 1,3- benzodioxol-5-yl, 2-methyl-l, 3-benzodioxol-5-yl, 2,2- dimethyl-1, 3-benzodioxol-5-yl, 2 , 2-dideutero-l, 3- benzodioxol-5-yl, 2, 2-difluoro-1, 3-benzodioxol-5-yl, l,4-benzodioxan-6-yl, 5-benzimidazolyl, 2- (methoxycarbonylamino)benzimidazol-5-yl, 6-quinolinyl, 7-quinolinyl, 6-isoquinolinyl or 7-isoquinolinyl radicals; more preferably, R represents phenyl, 2- naphthyl, 4-methoxyphenyl, 4-hydroxyphenyl, benzothiazol-5-yl, benzothiazol-6-yl, benzoxazol-5-yl, 2,3-dihydrobenzofuran-5-yl, benzofuran-5-yl, 1,3- benzodioxol-5-yl, 2-methyl-l, 3-benzodioxol-5-yl, 2,2- dimethyl-1, 3-benzodioxol-5-yl , 2 , 2-dideutero-l, 3- benzodioxol-5-yl, 2 , 2-difluoro-1, 3-benzodioxol-5-yl, 1, 4-benzodioxan-6-yl, 2- (methoxycarbonylamino) benzothiazol-5-yl, 2-
(methoxycarbonylamino)benzothiazol-6-yl or 2- (methoxycarbonylamino)benzimidazol-5-yl radicals; and most preferably, R4 represents phenyl, 4-methoxyphenyl, 4-hydroxyphenyl, benzothiazol-5-yl, benzothiazol-6-yl, 2,3-dihydrobenzofuran-5-yl, benzofuran-5-yl, 1,3- benzodioxol-5-yl, 2-methyl-l, 3-benzodioxol-5-yl, 2,2- dimethyl-1, 3-benzodioxol-5-yl, 2 , 2-dideutero-l, 3- benzodioxol-5-yl, 2 , 2-difluoro-1, 3-benzodioxol-5-yl, l,4-benzodioxan-6-yl, 2- (methoxycarbonylamino) benzothiazol-6-yl or 2-
(methoxycarbonylamino)benzimidazol-5-yl radicals;
R10 represents hydrogen, alkyl, hydroxyalkyl or alkoxyalkyl radicals, wherein alkyl is 1-3 carbon atoms; preferably R represents a hydrogen radical;
R represents alkyl radical of 1-5 carbon atoms, preferably methyl, isopropyl, butyl, secbutyl or isobutyl radicals; hydroxyalkyl radical of 1-4 carbon atoms, preferably hydroxymethyl or hydroxyethyl radicals; alkoxyalkyl radical of 1-4 alkyl carbon atoms, preferably methoxymethyl or methoxyethyl radicals; or hydrogen, benzyl, imidazolylmethyl, -CH2CH2CONH2, -CH2CONH2, -CH2CH2SCH3 or -CHSCH3 radicals or the sulfone or sulfoxide derivatives thereof; more preferably R11 represents a hydrogen radical;
R12 represents hydrogen, hydroxyalkyl or alkoxyalkyl radicals; preferably, R12 represents hydrogen, hydroxyalkyl or alkoxyalkyl radicals, wherein alkyl is 1-3 carbon atoms; preferably R12 represents a hydrogen radical; and
R13 and R14 each independently represent hydrogen, hydroxy, alkoxy, hydroxyalkoxy, hydroxyalkyl or alkoxyalkyl radicals; preferably, R13 and R each independently represent hydrogen, hydroxy, alkoxy, 2- hydroxyethoxy, hydroxyalkyl or alkoxyalkyl radicals, wherein alkyl is 1-3 carbon atoms; more preferably, R and R14 each independently represent hydrogen, hydroxy, methoxy or ethoxy radicals; or
R12 and R13 or R13 and R14 along with the carbon atoms to which they are attached represent 5-6 ring membered heteroaryl or benzo radical, each of which is optionally substituted with at least one hydroxy or alkoxy radical of 1-3 carbon atoms; preferably R 12 and R13 or R13 and R14 along with the carbon atoms to which they are attached represent benzo radical, which is optionally substituted with at least one hydroxy or methoxy radical.
The absolute stereochemistry of the carbon atom of -CH(OH)- group is preferably (R) . The absolute stereochemistry of the carbon atom of -CH(R1)- group is preferably (S) . The absolute stereochemistry of the carbon atom of-CH(R2)- groups is preferably (S) .
A family of compounds of particular interest within Formula I are compounds embraced by the formula
Figure imgf000013_0001
or a pharmaceutically acceptable salt, prodrug or ester thereof, wherein n, R1, R2, R3, and R4 are as defined above.
A family of compounds of further interest within Formula II are compounds embraced by the formula
Figure imgf000013_0002
or a pharmaceutically acceptable salt, prodrug or ester thereof, wherein R1, R2 , R3, R4 and n are as defined above.
A more preferred family of compounds within Formula III consists of compounds or a pharmaceutically acceptable salt, prodrug or ester thereof, wherein n represents 1;
R1 represents sec-butyl, tert-butyl, iso-propyl, 3- propynyl or -C(CH3) 2S(0) 2CH3 radicals; R2 represents a benzyl radical;
R3 represents propyl, isoa yl, isobutyl, butyl, cyclohexyl, cycloheptyl, cyclopentylmethyl or cyclohexylmethyl radicals; and
R4 is as defined above,
Compounds of interest include the following:
2S-[ [ (pyrrolidin-1-yl) acetyl] amino] -N-[2R-hydroxy-3- [ [ (l,3-benzodioxol-5-yl)sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -3 , 3- dimethyl-butanamide;
2S- [ [ (pyrrolidin-l-yl) acetyl] amino] -N- [2R-hydroxy-3- [ [ (l,3-benzodioxol-5-yl)sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -3-methyl- butanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -N-[2R-hydroxy-3-
[[ (l,3-benzodioxol-5-yl)sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -3S-methyl- pentanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -N- [2R-hydroxy-3- [ [ (l,3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -4- pentynamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -N- [2R-hydroxy-3- [ [phenylsulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -3 , 3-dimethyl-butanamide; S-[ [ (pyrrolidin-1-yl) acetyl] amino]-N-[2R-hydroxy-3- [phenylsulfonyl] (2-methylpropyl) amino] -1S- phenylmethyl)propyl] -3-methyl-butanamide;
s~[ [ (pyrrolidin-l-yl) acetyl] amino] -N-[2R-hydroxy-3- [phenylsulfonyl] (2-methylpropyl) amino] -1S- phenylmethyl) propyl] -3S-methyl-pentanamide;
s~ [ [ (pyrrolidin-l-yl) acetyl] amino] -N- [2R-hydroxy-3- r [phenylsulfonyl] (2-methylpropyl) amino] -LS- phenylmethyl) propyl] -4-pentynamide;
s~[ C (pyrrolidin-l-yl) acetyl] amino] -N-[2R-hydroxy-3- [ (4-methoxyphenyl-sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -3 , 3-dimethyl-butanamide;
s~[ [ (pyrrolidin-l-yl) acetyl] amino] -N-[2R-hydroxy-3- [ (4-methoxyphenyl) sulfonyl] (2-methylpropyl) amino] -1S- phen l ethy1) propyl] -3-methyl-butanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -N-[2R-hydroxy-3- [ (4-methoxyphenyl) sulfonyl] (2-methylpropyl) amino] -1S- phenylmethy1) propyl] -3S-methyl-pentanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl]amino]-N-[2R-hydroxy-3-
[ (4-methoxyphenyl) sulfonyl] (2-methylpropyl) amino] -1S- phenylmethy1) propyl] -4-pentynamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino]-N-[2R-hydroxy-3- [[ (2,3-dihydrobenzofuran-5-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl] -3 , 3-dimethyl- butana ide; 2S-[ [ (pyrrolidin-l-yl) acetyl]amino]-N-[2R-hydroxy-3- [ [ (2, 3-dihydrobenzofuran-5-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl] -3-methyl-butanamide;
2S-[[ (pyrrolidin-l-yl) acetyl] amino] -N-[2R-hydroxy-3-
[ [ (2 , 3-dihydrobenzofuran-5-yl) sulfonyl] (2-methylpropyl) amino] -IS-phenylmethyl) propyl] -3S-methyl-pentanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino] )N-[2R-hydroxy-3- [ [ (2,3-dihydrobenzofuran-5-yl)sulfonyl] 2-methylpropyl) amino] -lS-phenylmethy1) propyl] -4-pentynamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino]-N-[2R-hydroxy-3- [ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -3 , 3-dimethyl-butanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl]amino]-N-[2R-hydroxy-3- [ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -3-methyl-butanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl]amino]-N-[2R-hydroxy-3- [ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -3S-methyl-pentanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -N-[2R-hydroxy-3- [ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - 1S- (phenylmethyl) propyl] -4-pentynamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino]-N-[2R-hydroxy-3- [[ (2-naphthyl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl] -3 , 3-dimethyl-butanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl]amino] -N- [2R-hydroxy-3- [ [ (2-naphthyl) sulfonyl] (2-methylpropyl)amino]-lS- (phenylmethyl) propyl] -3-methyl-butanamide; 2S-[ [ (pyrrolidin-l-yl) acetyl] amino]-N-[2R-hydroxy-3- [[ (2-naphthyl) sulfonyl] (2-methylpropyl) amino]-1S- (phenylmethyl) propyl] -3S-methyl-pentanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -N-[2R-hydroxy-3- [ [ (2-naphthyl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -4-pentynamide;
2S-[ [ (pyrrolidin-l-yl) acetyl] amino]-N-[2R-hydroxy-3- t [ (l,4-benzodioxan-6-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -3 , 3- dimethy1-butanamide;
2S-[[ (pyrrolidin-l-yl) acetyl] amino]-N-[2R-hydroxy-3- [ [ (l,4-benzodioxan-6-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -3-methyl- butanamide;
2S-[ [ (pyrrolidin-l-yl) acetyl]amino]-N-[2R-hydroxy-3- [ [ (l,4-benzodioxan-6-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -3S-methyl- pentanamide; and
2S-[ [ (pyrrolidin-l-yl) acetyl]amino] -N-[2R-hydroxy-3- [ [ (1, 4-benzodioxan-6-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -4- pentynamide.
As utilized herein, the term "alkyl", alone or in combination, means a straight-chain or branched-chain alkyl radical containing preferably from 1 to 8 carbon atoms, more preferably from 1 to 5 carbon atoms, most preferably 1 to 3 carbon atoms. Examples of such radicals include methyl, ethyl, n-propyl, isopropyl, n- butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso- amyl, hexyl, octyl and the like. The term "alkenyl", alone or in combination, means a straight-chain or branched-chain hydrocarbon radical having one or more double bonds and containing preferably from 2 to 10 carbon atoms, more preferably from 2 to 8 carbon atoms, most preferably from 2 to 5 carbon atoms. Examples of suitable alkenyl radicals include ethenyl, propenyl, 2- methylpropenyl, 1,4-butadienyl and the like. The term "alkynyl", alone or in combination, means a straight- chain or branched chain hydrocarbon radical having one or more triple bonds and containing preferably from 2 to 10 carbon atoms, more preferably from 2 to 5 carbon atoms. Examples of alkynyl radicals include ethynyl, propynyl (propargyl) , butynyl and the like. The term
"alkoxy", alone or in combination, means an alkyl ether radical wherein the term alkyl is as defined above. Examples of suitable alkyl ether radicals include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso- butoxy, sec-butoxy, tert-butoxy and the like. The term "cycloalkyl", alone or in combination, means a saturated or partially saturated monocyclic, bicyclic or tricyclic alkyl radical wherein each cyclic moiety contains preferably from 3 to 8 carbon atom ring members, more preferably from 3 to 7 carbon atom ring members, most preferably from 5 to 6 carbon atom ring members, and which may optionally be a benzo fused ring system which is optionally substituted as defined herein with respect to the definition of aryl. Examples of such cycloalkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, octahydronaphthyl, 2,3-dihydro- lH-indenyl, ada antyl and the like. "Bicyclic" and "tricyclic" as used herein are intended to include both fused ring systems, such as naphthyl and β-carbolinyl, and substituted ring systems, such as biphenyl, phenylpyridyl, naphthyl and diphenylpiperazinyl. The term "cycloalkylalkyl" means an alkyl radical as defined above which is substituted by a cycloalkyl radical as defined above. Examples of such cycloalkylalkyl radicals include cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, 1-cyclopentylethyl, 1-cyclohexylethyl, 2-cyclopentylethyl, 2- cyclohexylethyl, cyclobutylpropyl, cyclopentylpropyl, cyclohexylbutyl and the like. The term "benzo", alone or in combination, means the divalent radical C6H4= derived from benzene. The term "aryl", alone or in combination, means a phenyl or naphthyl radical which is optionally substituted with one or more substituents selected from alkyl, alkoxy, halogen, hydroxy, amino, nitro, cyano, haloalkyl, carboxy, alkoxycarbonyl, cycloalkyl, heterocyclo, alkanoylamino, amido, amidino, alkoxycarbonylamino, N-alkylamidino, alkylamino, dialkylamino, N-alkylamido, N,N-dialkylamido, aralkoxycarbonylamino, alkylthio, alkylsulfinyl, alkylsulfonyl and the like. Examples of aryl radicals are phenyl, p-tolyl, 4-methoxyphenyl, 4-(tert- butoxy) phenyl, 3-methyl-4-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 3-nitrophenyl, 3-aminophenyl, 3- acetamidophenyl, 4-acetamidophenyl, 2-methyl-3- acetamidophenyl, 4-CF3-phenyl, 2-methyl-3-aminophenyl, 3-methy1-4-aminopheny1, 2-amino-3- methylphenyl , 2,4-dimethyl-3-aminophenyl, 4- hydroxyphenyl, 3-methyl-4-hydroxyphenyl, 1-naphthyl, 2- naphthyl, 3-amino-l-naphthyl, 2-methyl-3-amino-l- naphthyl, 6-amino-2-naphthyl, 4 , 6-dimethoxy-2-naphthyl, piperazinylphenyl and the like. The terms "aralkyl" and "aralkoxy", alone or in combination, means an alkyl or alkoxy radical as defined above in which at least one hydrogen atom is replaced by an aryl radical as defined above, such as benzyl, benzyloxy, 2-phenylethyl, dibenzylmethyl, hydroxyphenylmethyl, methylphenylmethy1 , diphenylmethyl, diphenylmethoxy, 4-methoxyphenylmethoxy and the like. The term "aralkoxycarbonyl" , alone or in combination, means a radical of the formula aralkyl-O- C(0)- in which the term "aralkyl" has the significance given above. Examples of an aralkoxycarbonyl radical are benzyloxycarbonyl and 4- methoxyphenylmethoxycarbonyl . The term "aryloxy" means a radical of the formula aryl-O- in which the term aryl has the significance given above. The term "alkanoyl" , alone or in combination, means an acyl radical derived from an alkanecarboxylic acid, examples of which include acetyl, propionyl, butyryl, valeryl, 4-methylvaleryl, and the like. The term "cycloalkylcarbonyl" means an acyl radical of the formula cycloalkyl-C(O) - in which the term "cycloalkyl" has the significance give above, such as cyclopropylcarbonyl, cyclohexylcarbonyl, adamantylcarbonyl, 1, 2 , 3 ,4-tetrahydro-2-naphthoyl, 2- acetamido-1, 2,3, 4-tetrahydro-2-naphthoyl, 1-hydroxy- l,2,3,4-tetrahydro-6-naphthoyl and the like. The term
"aralkanoyl" means an acyl radical derived from an aryl- substituted alkanecarboxylic acid such as phenylacetyl, 3-phenylpropionyl (hydrocinnamoyl) , 4-phenylbutyryl, (2- naphthyl) acetyl, 4-chlorohydrocinnamoyl, 4- aminohydrocinnamoyl, 4-methoxyhydrocinnamoyl, and the like. The term "aroyl" means an acyl radical derived from an arylcarboxylic acid, "aryl" having the meaning given above. Examples of such aroyl radicals include substituted and unsubstituted benzoyl or napthoyl such as benzoyl, 4-chlorobenzoyl, 4-carboxybenzoyl, 4-
(benzyloxycarbonyl) benzoyl, 1-naphthoyl, 2-naphthoyl, 6- carboxy-2-naphthoyl, 6- (benzyloxycarbonyl) -2-naphthoyl, 3-benzyloxy-2-naphthoyl, 3-hydroxy-2-naphthoyl, 3- (benzyloxyforma ido) -2-naphthoyl, and the like. The term "heterocyclo," alone or in combination, means a saturated or partially unsaturated monocyclic, bicyclic or tricyclic heterocycle radical containing at least one, preferably 1 to 4 , more preferably 1 to 2, nitrogen, oxygen or sulfur atom ring members and having preferably 3 to 8 ring members in each ring, more preferably 3 to 7 ring members in each ring and most preferably 5 to 6 ring members in each ring. "Heterocyclo" is intended to include sulfones, sulfoxides, N-oxides of tertiary nitrogen ring members, and carbocyclic fused and benzo fused ring systems.
Such heterocyclo radicals may be optionally substituted on at least one, preferably 1 to 4 , more preferably 1 to 2, carbon atoms by halogen, alkyl, alkoxy, hydroxy, oxo, aryl, aralkyl, heteroaryl, heteroaralkyl, amidino, N- alkylamidino, alkoxycarbonylammo, alkylsulfonylammo and the like, and/or on a secondary nitrogen atom (i.e., -NH-) by hydroxy, alkyl, aralkoxycarbonyl, alkanoyl, heteroaralkyl, phenyl or phenylalkyl, and/or on a tertiary nitrogen atom (i.e., =N-) by oxido. "Heterocycloalkyl" means an alkyl radical as defined above in which at least one hydrogen atom is replaced by a heterocyclo radical as defined above, such as pyrrolidinylmethy1, tetrahydrothienyl ethyl, pyridylmethyl and the like. The term "heteroaryl", alone or in combination, means an aromatic heterocyclo radical as defined above, which is optionally substituted as defined above with respect to the definitions of aryl and heterocyclo. Examples of such heterocyclo and heteroaryl groups are pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiamorpholinyl, pyrrolyl, imidazolyl (e.g., imidazol 4-yl, 1- benzyloxycarbonylimidazol-4-yl, etc.), pyrazolyl, pyridyl, (e.g., 2- (l-piperidinyl)pyridyl and 2-(4-benzyl piperazin-1-yl-l-pyridinyl, etc.), pyrazinyl, pyrimidinyl, furyl, tetrahydrofuryl, thienyl, tetrahydrothienyl and its sulfoxide and sulfone derivatives, triazolyl, oxazolyl, thiazolyl, indolyl (e.g., 2-indolyl, etc.), quinolinyl, (e.g., 2- quinolinyl, 3-quinolinyl, l-oxido-2-quinolinyl, etc.), isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl, etc.), tetrahydroquinolinyl (e.g., 1,2 , 3 ,4-tetrahydro-2- quinolyl, etc.), 1,2 , 3 ,4-tetrahydroisoquinolinyl (e.g., 1,2,3, 4-tetrahydro-l-oxo-isoquinolinyl , etc . ) , quinoxalinyl, β-carbolinyl, 2-benzofurancarbonyl, l-,2- ,4- or 5-benzimidazolyl, methylenedioxyphen-4-yl, methylenedioxyphen-5-yl , ethylenedioxyphenyl , benzothiazolyl, benzopyranyl, benzofuryl, 2,3- dihydrobenzofuryl, benzoxazolyl, thiophenyl and the like. The term "cycloalkylalkoxycarbonyl" means an acyl group derived from a cycloalkylalkoxycarboxylic acid of the formula cycloalkylalkyl-O-COOH wherein cycloalkylalkyl has the meaning given above. The term "aryloxyalkanoyl" means an acyl radical of the formula aryl-O-alkanoyl wherein aryl and alkanoyl have the meaning given above. The term
"heterocycloalkoxycarbonyl" means an acyl group derived from heterocycloalkyl-O-COOH wherein heterocycloalkyl is as defined above. The term "heterocycloalkanoyl" is an acyl radical derived from a heterocycloalkylcarboxylic acid wherein heterocyclo has the meaning given above. The term "heterocycloalkoxycarbonyl" means an acyl radical derived from a heterocycloalkyl-O-COOH wherein heterocyclo has the meaning given above. The term "heteroaryloxycarbonyl" means an acyl radical derived from a carboxylic acid represented by heteroaryl-O-COOH wherein heteroaryl has the meaning given above. The term "a inocarbonyl" alone or in combination, means an amino-substituted carbonyl (carbamoyl) group wherein the amino group can be a primary, secondary or tertiary amino group containing substituents selected from alkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl radicals and the like. The term "aminoalkanoyl" means an acyl group derived from an amino-substituted alkylcarboxylic acid wherein the amino group can be a primary, secondary or tertiary amino group containing substituents selected from alkyl, aryl, aralkyl, cycloalkyl, cycloalkylalkyl radicals and the like. The term "halogen" means fluorine, chlorine, bromine or iodine. The term "haloalkyl" means an alkyl radical having the meaning as defined above wherein one or more hydrogens are replaced with a halogen. Examples of such haloalkyl radicals include chloromethyl, 1-bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1, 1, 1-trifluoroethyl and the like. The term "leaving group" (L or W) generally refers to groups readily displaceable by a nucheophile( such as an a ine, a thiol or an alcohol nucleophile. Such leaving groups are well known in the art. Examples of such leaving groups include, but are not limited to, N-hydroxysuccinimide, N- hydroxybenzotriazole, halides, triflates, tosylates and the like. Preferred leaving groups are indicated herein where appropriate.
Procedures for preparing the compounds of Formula I are set forth below. It should be noted that the general procedure is shown as it relates to preparation of compounds having the specified stereochemistry, for example, wherein the absolute stereochemistry about the hydroxyl group is designated as (R) . However, such procedures are generally applicable to those compounds of opposite configuration, e.g., where the stereochemistry about the hydroxyl group is (S) . In addition, the compounds having the (R) stereochemistry can be utilized to produce those having the (S) stereochemistry. For example, a compound having the (R) stereochemistry can be inverted to the (S) stereochemistry using well-known methods.
Preparation of Compounds of Formula I The compounds of the present invention represented by Formula I above can be prepared utilizing the following general procedures as schematically shown in Schemes I and II.
SCHEME I
Figure imgf000025_0001
a) deprotection; X = Cl or Br, SCHEME II
Figure imgf000026_0001
a) deprotection; X = Cl or Br; L = leaving group. SCHEME IIA
Figure imgf000027_0001
Figure imgf000027_0002
Figure imgf000027_0003
An N-protected chloroketone derivative of an amino acid having the formula:
Figure imgf000028_0001
wherein P represents an amino protecting group, and R2 is as defined above, is reduced to the corresponding alcohol utilizing an appropriate reducing agent. Suitable amino protecting groups are well known in the art and include carbobenzoxy, t-butoxycarbonyl, and the like. A preferred amino protecting group is carbobenzoxy. A preferred N-protected chloroketone is N-benzyloxycarbonyl-L-phenylalanine chloromethyl ketone. A preferred reducing agent is sodium borohydride. The reduction reaction is conducted at a temperature of from -10°C to about 25°C, preferably at about 0°C, in a suitable solvent system such as, for example, tetrahydrofuran, and the like. The N-protected chloroketones are commercially available, e.g., such as from Bachem, Inc., Torrance, California. Alternatively, the chloroketones can be prepared by the procedure set forth in S. J. Fittkau, J. Prakt. Chem. , 315, 1037 (1973) , and subsequently N-protected utilizing procedures which are well known in the art.
The halo alcohol can be utilized directly, as described below, or, preferably, is reacted, preferably at room temperature, with a suitable base in a suitable solvent system to produce an N-protected amino epoxide of the formula:
Figure imgf000029_0001
wherein P and R are as defined above. Suitable solvent systems for preparing the amino epoxide include ethanol, methanol, isopropanol, tetrahydrofuran, dioxane, and the like including mixtures thereof. Suitable bases for producing the epoxide from the reduced chloroketone include potassium hydroxide, sodium hydroxide, potassium t-butoxide, DBU and the like. A preferred base is potassium hydroxide.
Alternatively, a protected amino epoxide can be prepared, such as in co-owned and co-pending PCT Patent Application Serial No. PCT/US93/04804 (WO 93/23388) and PCT/US94/12201, and US Patent Application Attorney
Docket No. C-2860, each of which is incorporated herein by reference in their entirety) disclose methods of preparing chiral epoxide, chiral cyanohydrin, chiral amine and other chiral intermediates useful in the preparation of retroviral protease inhibitors, starting with a DL-, D- or L-amino acid which is reacted with a suitable amino-protecting group in a suitable solvent to produce an amino-protected amino acid ester. For the purposes of illustration, a protected L-amino acid with the following formula will be used to prepare the inhibitors of this invention:
Figure imgf000029_0002
wherein P represents carboxyl-protecting group, e.g., methyl, ethyl, benzyl, tertiary-butyl, 4- methoxyphenylmethy1 and the like; R2 is as defined above; and P 1 and P2 i•ndependently are selected from amine protecting groups, including but not limited to, aralkyl, substituted aralkyl, cycloalkenylalkyl and substituted cycloalkenylaloyl, allyl, substituted allyl, acyl, alkoxycarbonyl, aralkoxycarbonyl and silyl. Examples of aralkyl include, but are not limited to benzyl, ortho-methylbenzyl, trityl and benzhydryl, which can be optionally substituted with halogen, alkyl of Cι~ Cg, alkoxy, hydroxy, nitro, alkylene, amino, alkylamino, acylamino and acyl, or their salts, such as phosphonium and ammonium salts. Examples of aryl groups include phenyl, naphthalenyl, indanyl, anthracenyl, durenyl, 9- (9-phenylfluorenyl) and phenanthrenyl, cycloalkenylalkyl or substituted cycloalkylenylalkyl radicals containing cycloalkyls of Cg-Cio- Suitable acyl groups include carbobenzoxy, t-butoxycarbonyl, iso-butoxycarbonyl, benzoyl, substituted benzoyl, butyryl, acetyl, tri- fluoroacetyl, tri-chloroacetyl, phthaloyl and the like. Preferably P1 and P2 are independently selected from aralkyl and substituted aralkyl. More preferably, each of P1 and P2 is benzyl.
Additionally, the P1 and/or P2 protecting groups can form a heterocyclic ring with the nitrogen to which they are attached, for example, 1,2- bis(methylene) benzene, phthali idyl, succinimidyl, maleimidyl and the like and where these heterocyclic groups can further include adjoining aryl and cycloalkyl rings. In addition, the heterocyclic groups can be mono-, di- or tri-substituted, e.g., nitrophthalimidyl. The term silyl refers to a silicon atom optionally substituted by one or more alkyl, aryl and aralkyl groups.
Suitable silyl protecting groups include, but are not limited to, trimethylsilyl, triethylsilyl, tri- isopropylsilyl , tert-butyldimethylsilyl, dimethylphenylsilyl, 1,2-bis (dimethylsilyl) benzene, 1,2- bis(dimethylsilyl) ethane and diphenylmethylsilyl. Silylation of the amine functions to provide mono- or bis-disilylamine can provide derivatives, of the aminoalcohol, amino acid, amino acid esters and amino acid amide. In the case of amino acids, amino acid esters and amino acid amides, reduction of the carbonyl function provides the required mono- or bis-silyl aminoalcohol. Silylation of the aminoalcohol can lead to the N,N,0-tri-silyl derivative. Removal of the silyl function from the silyl ether function is readily accomplished by treatment with, for example, a metal hydroxide or ammonium flouride reagent, either as a discrete reaction step or in situ during the preparation of the amino aldehyde reagent. Suitable silylating agents are, for example, trimethylsilyl chloride, tert- buty-dimethylsilyl chloride, phenyldimethylsilyl chloride, diphenylmethylsilyl chloride or their combination products with imidazole or DMF. Methods for silylation of amines and removal of silyl protecting groups are well known to those skilled in the art. Methods of preparation of these amine derivatives from corresponding amino acids, amino acid amides or amino acid esters are also well known to those skilled in the art of organic chemistry including amino acid/amino acid ester or aminoalcohol chemistry.
The amino-protected L-amino acid ester is then reduced, to the corresponding alcohol. For example, the amino-protected L-amino acid ester can be reduced with diisobutylaluminum hydride at -78 °C in a suitable solvent such as toluene. Preferred reducing agents include lithium aluminium hydride, lithium borohydride, sodium borohydride, borane, lithium tri-ter- butoxyaluminum hydride, borane/THF complex. Most preferably, the reducing agent is diisobutylaluminum hydride (DiBAL-H) in toluene. The resulting alcohol is then converted, for example, by way of a Swern oxidation, to the corresponding aldehyde, of the formula:
R2
Figure imgf000032_0001
wherein P1, P2 and R2 are as defined above. Thus, a dichloromethane solution of the alcohol is added to a cooled (-75 to -68 °C) solution of oxalyl chloride in dichloromethane and DMSO in dichloromethane and stirred for 35 minutes.
Acceptable oxidizing reagents include, for example, sulfur trioxide-pyridine complex and DMSO, oxalyl chloride and DMSO, acetyl chloride or anhydride and DMSO, trifluoroacetyl chloride or anhydride and DMSO, methanesulfonyl chloride and DMSO or tetrahydro thiaphene-S-oxide, toluenesulfonyl bromide and DMSO, trifluoromethanesulfonyl anhydride (triflic anhydride) and DMSO, phosphorus pentachloride and DMSO, dimethylphosphoryl chloride and DMSO and isobutyl chloroformate and DMSO. The oxidation conditions reported by Reetz et al (An ew Che . , 99, p. 1186,
(1987), Anqew Chem. Int. Ed. En l., 26, p. 1141, 1987) employed oxalyl chloride and DMSO at -78°C. The preferred oxidation method described in this invention is sulfur trioxide pyridine complex, triethylamine and DMSO at room temperature. This system provides excellent yields of the desired chiral protected amino aldehyde usable without the need for purification i.e., the need to purify kilograms of intermediates by chromatography is eliminated and large scale operations are made less hazardous. Reaction at room temperature also eliminated the need for the use of low temperature reactor which makes the process more suitable for commercial production.
The reaction may be carried out under an inert atmosphere such as nitrogen or argon, or normal or dry air, under atmospheric pressure or in a sealed reaction vessel under positive pressure. Preferred is a nitrogen atmosphere. Alternative amine bases include, for example, tri-butyl amine, tri-isopropyl amine, N- methylpiperidine, N-methyl morpholine, azabicyclononane, diisopropylethylamine, 2,2,6, 6-tetramethylpiperidine, N,N-dimethylaminopyridine, or mixtures of these bases. Triethylamine is a preferred base. Alternatives to pure DMSO as solvent include mixtures of DMSO with non-protic or halogenated solvents such as tetrahydrofuran, ethyl acetate, toluene, xylene, dichloromethane, ethylene dichloride and the like. Dipolar aprotic co-solvents include acetonitrile, dimethylformamide, dimethylacetamide, acetamide, tetramethyl urea and its cyclic analog, N-methylpyrrolidone, sulfolane and the like. Rather than N,N-dibenzylphenylalaninol as the aldehyde precursor, the phenylalaninol derivatives discussed above can be used to provide the corresponding N-monosubstituted (either P1 or P2 = H) or N,N- disubstituted aldehyde. In addition, hydride reduction of an amide or ester derivative of the corresponding benzyl (or other suitable protecting group) nitrogen protected phenylalanme, substituted phenylalanme or cycloalkyl analog of phenylalanine derivative can be carried out to provide the aldehydes. Hydride transfer is an additional method of aldehyde synthesis under conditions where aldehyde condensations are avoided, cf, Oppenauer Oxidation.
The aldehydes of this process can also be prepared by methods of reducing protected phenylalanine and phenylalanine analogs or their amide or ester derivatives by, e.g., sodium amalgam with HC1 in ethanol or lithium or sodium or potassium or calcium in ammonia. The reaction temperature may be from about -20°C to about 45°C, and preferably from abut 5°C to about 25°C. Two additional methods of obtaining the nitrogen protected aldehyde include oxidation of the corresponding alcohol with bleach in the presence of a catalytic amount of 2 , 2, 6, 6-tetramethyl-l-pyridyloxy free radical. In a second method, oxidation of the alcohol to the aldehyde is accomplished by a catalytic amount of tetrapropylammonium perruthenate in the presence of N-methylmorpholine-N-oxide.
Alternatively, an acid chloride derivative of a protected phenylalanine or phenylalanine derivative as disclosed above can be reduced with hydrogen and a catalyst such as Pd on barium carbonate or barium sulphate, with or without an additional catalyst moderating agent such as sulfur or a thiol (Rosenmund Reduction) . The aldehyde resulting from the Swern oxidation is then reacted with a halomethyllithium reagent, which reagent is generated in situ by reacting an alkyllithium or arylithium compound with a dihalomethane represented by the formula X1CH2X2 wherein X1 and X2 independently represent I, Br or Cl. For example, a solution of the aldehyde and chloroiodomethane in THF is cooled to -78°C and a solution of n-butyllithium in hexane is added. The resulting product is a mixture of diastereomers of the corresponding amino-protected epoxides of the formulas:
Figure imgf000035_0001
The diastereomers can be separated e.g., by chromatography, or, alternatively, once reacted in subsequent steps the diastereomeric products can be separated. A D-amino acid can be utilized in place of the L-amino acid in order to prepare compounds having an (S) stereochemistry at the carbon bonded to R .
The addition of chloromethylithium or bromomethylithium to a chiral amino aldehyde is highly diastereoselective. Preferably, the chloromethyllithium or bromomethylithium is generated in-situ from the reaction of the dihalomethane and n-butyllithium. Acceptable methyleneating halomethanes include chloroiodomethane, bromochloromethane, dibromomethane, diiodomethane, bromofluoromethane and the like. The sulfonate ester of the addition product of, for example, hydrogen bromide to formaldehyde is also a methyleneating agent. Tetrahydrofuran is the preferred solvent, however alternative solvents such as toluene, dimethoxyethane, ethylene dichloride, methylene chloride can be used as pure solvents or as a mixture. Dipolar aprotic solvents such as acetonitrile, DMF, N- methylpyrrolidone are useful as solvents or as part of a solvent mixture. The reaction can be carried out under an inert atmosphere such as nitrogen or argon. For n- butyl lithium can be substituted other organometalic reagents reagents such as methyllithium, tert-butyl lithium, sec-butyl lithium, phenyllithium, phenyl sodium and the like. The reaction can be carried out at temperatures of between about -80°C to 0°C but preferably between about -80°C to -20°C. The most preferred reaction temperatures are between -40°C to - 15°C. Reagents can be added singly but multiple additions are preferred in certain conditions. The preferred pressure of the reaction is atmospheric however a positive pressure is valuable under certain conditions such as a high humidity environment.
Alternative methods of conversion to the epoxides of this invention include substitution of other charged methylenation precurser species followed by their treatment with base to form the analogous anion. Examples of these species include trimethylsulfoxonium tosylate or triflate, tetramethylammonium halide, methyldiphenylsulfoxonium halide wherein halide is chloride, bromide or iodide.
The conversion of the aldehydes of this invention into their epoxide derivative can also be carried out in multiple steps. For example, the addition of the anion of thioanisole prepared from, for example, a butyl or aryl lithium reagent, to the protected aminoaldehyde, oxidation of the resulting protected aminosulfide alcohol with well known oxidizing agents such as hydrogen peroxide, tert-butyl hypochlorite, bleach or sodium periodate to give a sulfoxide. Alkylation of the sulfoxide with, for example, methyl iodide or bromide, methyl tosylate, methyl mesylate, methyl triflate, ethyl bromide, isopropyl bromide, benzyl chloride or the like, in the presence of an organic or inorganic base Alternatively, the protected aminosulfide alcohol can be alkylated with, for example, the alkylating agents above, to provide a sulfonium salts that are subsequently converted into the subject epoxides with tert-amine or mineral bases.
The desired epoxides formed, using most preferred conditions, diastereoselectively in ratio amounts of at least about an 85:15 ratio (S:R) . The product can be purified by chromatography to give the diastereomerically and enantiomerically pure product but it is more conveniently used directly without purification to prepare retroviral protease inhibitors. The foregoing process is applicable to mixtures of optical isomers as well as resolved compounds. If a particular optical isomer is desired, it can be selected by the choice of starting material, e.g., L- phenylalanine, D-phenylalanine, L-phenylalaninol, D- phenylalaninol, D-hexahydrophenylalaninol and the like, or resolution can occur at intermediate or final steps. Chiral auxiliaries such as one or two equivalents of camphor sulfonic acid, citric acid, camphoric acid, 2- methoxyphenylacetic acid and the like can be used to form salts, esters or amides of the compounds of this invention. These compounds or derivatives can be crystallized or separated chromatographically using either a chiral or achiral column as is well known to those skilled in the art. The amino epoxide is then reacted, in a suitable solvent system, with an equal amount, or preferably an excess of, a desired amine of the formula R NH , wherein R is hydrogen or is as defined above. The reaction can be conducted over a wide range of temperatures, e.g., from about 10°C to about 100°C, but is preferably, but not necessarily, conducted at a temperature at which the solvent begins to reflux. Suitable solvent systems include protic, non-protic and dipolar aprotic organic solvents such as, for example, those wherein the solvent is an alcohol, such as methanol, ethanol, isopropanol, and the like, ethers such as tetrahydrofuran, dioxane and the like, and toluene, N,N-dimethylformamide, dimethyl sulfoxide, and mixtures thereof. A preferred solvent is isopropanol. The resulting product is a 3- (N-protected amino) -3-(R2) -1-(NHR3) -propan-2-ol derivative (hereinafter referred to as an amino alcohol) can be represented by the formulas:
Figure imgf000038_0001
wherein P, P1, P2, R2 and R3 are as described above. Alternatively, a haloalcohol can be utilized in place of the amino epoxide.
The amino alcohol defined above is then reacted in a suitable solvent with an activated sulfonyl function such as the sulfonyl chloride R4S02C1, the sulfonyl bromide RS02Br, the sulfonate ester or the corresponding sulfonyl anhydride, preferably in the presence of an acid scavenger. Suitable solvents in which the reaction can be conducted include methylene chloride, dichloromethane, ethyl acetate, tetrahydrofuran and the like. Suitable acid scavengers include triethylamine, pyridine as well as hindered amine bases such as diazabicyclooctane, diazabicycloundecene, 2,6 disubstituted pyridine derivatives, di-isopropylamine and the like. Inorganic bases such as sodium carbonate, potassium bicarbonate, calcium carbonate and the like may also be used if desired. The process of preparing the sulfonamide can be carried out as a discrete step following isolation of, for example, a sulfonyl chloride reagent or as part of a single pot process during the preparation of the activated sulfonyl function. Isolation of the sulfonyl chloride is preferred. The resulting sulfonamide derivative can be represented, depending on the epoxide utilized by the formulas
Figure imgf000039_0001
wherein P, P1, P2, R2, R3 and R4 are as defined above. These intermediates are useful for preparing inhibitor compounds of the present invention.
The sulfonyl halides of the formula R4S02X can be prepared by the reaction of a suitable aryl, heteroaryl and benzo fused heterocyclo Grignard or lithium reagents with sulfuryl chloride, or sulfur dioxide followed by oxidation with a halogen, preferably chlorine. Aryl, heteroaryl and benzo fused heterocyclo Grignard or lithium reagents can be prepared from their corresponding halide (such as chloro or bromo) compounds which are commercially available or readily prepared from commercially available starting materials using known methods in the art. Also, thiols may be oxidized to sulfonyl chlorides using chlorine in the presence of water under carefully controlled conditions. Additionally, sulfonic acids, such as arylsulfonic acids, heteroaryl and benzo fused heterocyclosulfonic acids, may be converted into sulfonyl halides using reagents such as PC15, S0C12, C1C(0)C(0)C1 and the like, and also to anhydrides using suitable dehydrating reagents. The sulfonic acids may in turn be prepared using procedures well known in the art. Some sulfonic acids are commercially available. In place of the sulfonyl halides, sulfinyl halides (RSOX) or sulfenyl halides (RSX) can be utilized to prepare compounds wherein the -S02- moiety is replaced by an -SO- or -S- moiety, respectively. Arylsulfonic acids, benzo fused heterocyclo sulfonic acids or heteroaryl sulfonic acids can be prepared by sulfonation of the aromatic ring by well known methods in the art, such as by reaction with sulfuric acid, S03, S03 complexes, such as DMF(S03) (also depicted as S03DMF, S03»DMF, DMFS03 and the like), pyridine (S03) , N,N-dimethylacetamide(S0 ) , and the like. Preferably, arylsulfonyl halides are prepared from aromatic compounds by reaction with DMF(S0 ) and S0C12 or
C1C(0)C(0)C1. Most preferably, benzo fused heterocyclo sulfonyl halides are prepared from aromatic compounds by reaction with DMF(S03) and CIC(O) C(O) Cl. The reactions may be performed step wise or in a single pot. A single pot reaction is understood to mean a single reaction system, reaction vessel, kettle, bomb, flask, and the like.
The use of the two pot process may be desirable when sulfonic acids are available commercially. Should the desired sulfonic, sulfinic or sulfenic acid or activated acid be commercially available, amidation of the amine intermediate discussed above and used to prepare, for example, the sulfonamide, may be carried out in either a one pot or two pot reaction or reaction series as indicated.
The reactions used in the method of converting a aromatic compound, including heteroaryl compounds and benzo fused heterocycles, into sulfonic,acids, activated sulfonic acids or sulfonamides is generally carried out under an inert atmosphere such as nitrogen, argon, or helium. The use of dry air is possible in some cases where it does not decrease the yield(s) or present a safety hazard. The solvent or solvent mixtures are water immiscible, for example, dichloroethane (ethylene dichloride) , methylene chloride, trichloroethane, toluene, tetrachloroethane and the like. A co-solvent may be used that is water miscible if desired. The temperatures of the various stages of the process or processes can vary depending upon reagents and other conditions. In general, the process exemplified, for example, by the preparation of 1,3-benzodioxolesulfonyl chloride is started by mixing reagents at room temperature although a range of about
0°C to about 45°C when S03 is used or to about 75°C when S03»DMF is used with dichloromethane. The S03«DMF reagent, with its higher melting point, is an especially desirable reagent allowing a range of reaction temperatures that can be controlled by rationing the amount of heat provided to the reaction vessel or the reflux temperature of the solvent(s). When analysis indicates that the conversion is as complete as desired, or at a later time if desired, the reaction can be worked up by methods know in the art, e.g., cooling, addition of water and/or an acid such as hydrochloric acid or sulfuric acid, to provide the sulfonic acid.
It may be chemically and economically desirable and/or efficient with respect to, for example, the use of chemist and equipment time, to prepare an intermediate such as the sulfonyl chloride by cooling the reaction mixture to a temperature of between about -25°C to about 65°C. The preferred range is between about 0°C and room temperature. The amount of cooling that is necessary will depend on the type of oxalyl halide that is added in the next step. When the reaction mixture is cooled, an oxalyl halide such as oxalyl chloride or oxalyl bromide can be added to the reaction vessel at such a rate as to control the reaction temperature. The temperature can range from about 0°C to about 50°C but preferably between about 0°C and room temperature. Most preferred is a range of between about 10°C and about 40°C. The reaction can be sampled for analysis. When analysis indicates that the conversion has been carried out, the excess reagents, if present, can be decomposed by the controlled addition of water. Care, such as that which would be expected of a trained person, is indicated in this operation. Isolation of the sulfonyl chloride proceeds by separation of the phases, washing, if desired, the aqueous phase with a water non-miscible solvent such as dichloroethane one to four times and drying the organic phases separately, in batches or as a single, combined solution. The drying agent can be a chemical such as magnesium sulfate or sodium sulfate or the water can be removed via an azeotrope. The solution remaining, which may or may not be mixed with various amounts of drying agent, may be filtered if desired. The solvent can be removed by distillation at atmospheric pressure or below or evaporation under reduced pressure using, for example, a rotary evaporator. It is also possible in some cases to crystallize the product at room temperature or below directly from the solvent or from a mixture of solvents such as, for example, ethylene dichloride and hexane, heptane, pentane, decane, cyclohexane, methylcyclohexane, decalin, toluene, xylene (mixed xylenes or pure xylenes) or the like. If a solid is obtained, it may be washed further if desired with, for example, hexane, and the dissolved product recovered otherwise it can be used directly in the next step. A crystallizing or washing process may provide product that is superior on an economic or scientific basis when used for the preparation of a intermediate substrate for catalytic processes such as reductive deprotection via removal of benzyl groups from an amine nitrogen. The isolated activated sulfonic acid product may be added directly as a solid or dissolved in a solvent prior to addition as a reagent to the next reaction. Isolation of, for example, a benzo fused heterocyclo sulfonyl chloride such as l,3-benzodioxole-5-sulfonyl chloride, prior to its reaction with an amine is preferred and the yield is high and may exceed 95%.
The conversion of the activated oxidized sulfur acid product into the sulfonamide, sulfinamide or sulfeneamide can be carried out in the same pot as its preparation if desired. Addition of the amine following reaction, decomposition or removal of the oxalyl chloride reagent with or without the addition of a hydrogen ion (H+) scavenger discussed above produces the amide product. The same product is obtained when the reaction is partially worked up by the addition of water as presented above. Again, the activated sulfur acid such as the sulfonyl chloride discussed above is used to form the sulfonamide with or without the addition of a proton scavenging agent. These conditions, for example, the Hinsberg Test, are know in the art. If desired, an excess of an amine intermediate of this invention can be used rather than one or more of the usual proton scavenging agents. The amine salt formed in this process can be isolated and reused according to the methods below. Should the product of, for example, the sulfonamide forming reaction is itself a basic amine, it may be isolated as a salt or as an amine.
Arylsulfonic acids, benzo fused heterocyclo sulfonic acids, heteroaryl sulfonic acids, arylmercaptans, benzo fused heterocyclo mercaptans, heteroarylmercaptans, arylhalides, benzo fused heterocyclo halides, heteroarylhalides, and the like are commercially available or can be readily prepared from starting materials commercially available using standard methods well known in the art. For example, a number of sulfonic acids (R4S03H) represented by the formulas
Figure imgf000044_0001
wherein A, B, Z, R6, R7 and R9 are as defined above, have been prepared from 1,2-benzenedithiol, 2- mercaptanphenol, 1, 2-benzenediol, 2-aminobenzothiazole, benzothiazole, 2-aminobenzimidazole, benzimidazole, and the like, which are commercially available, by Carter, US Patent 4,595,407; Ehrenfreund et al., US Patent 4,634,465; Yoder et al., J. Heterocycl. Chem. 4:166-167 (1967); Cole et al., Aust. J. Chem. 33:675-680 (1980); Cabiddu et al. , Synthesis 797-798 (1976); Ncube et al., Tet. Letters 2345-2348 (1978); Ncube et al., Tet. Letters 255-256 (1977) ; Ansink & Cerfontain, Rec. Trav. Chim.Pays-Bas 108:395-403 (1989) ; and Kajihara & Tsuchiya, EP 638564 Al, each of which are incorporated herein by reference in their entirety. For example, 1, 2-benzenedithiol, 2-mercaptanphenol or 1,2- benzenediol can be reacted with R6R7C(L')2, where L' is as defined below, preferably, Br or I, in the presence of a base, such as hydroxide, or R6R7C=Q in the presence of acid, such as toluenesulfonic acid, or P2Os., to prepare the substituted benzo fused heterocycle of formula
Figure imgf000045_0001
which can then be sulfonylated to the sulfonic acid above. For example, CF2Br2 or CD2Br2 can be reacted with 1, 2-benzenedithiol, 2-mercaptanphenol or 1,2- benzenediol in the presence of base to produce the compounds
Figure imgf000045_0002
respectively, wherein A and B are O or S and D is a deuterium atom. Also, when A and/or B represent S, the sulfur can be oxidized using the methods described below to the sulfone or sulfoxide derivatives.
Following preparation of the sulfonamide derivati •ve, the ami■no protecting group P or P1 and P2 amino protecting groups are removed under conditions which will not affect the remaining portion of the molecule. These methods are well known in the art and include acid hydrolysis, hydrogenolysis and the like. A preferred method involves removal of the protecting group, e.g., removal of a carbobenzoxy group, by hydrogenolysis utilizing palladium on carbon in a suitable solvent system such as an alcohol, acetic acid, and the like or mixtures thereof. Where the protecting group is a t-butoxycarbonyl group, it can be removed utilizing an inorganic or organic acid, e.g., HC1 or trifluoroacetic acid, in a suitable solvent system, e.g., dioxane or methylene chloride. The resulting product is the amine salt derivative.
Following neutralization of the salt, the amine is then coupled to the DL-, D-, or L-amino acid corresponding to the formula PNHCH(R1)C00H, wherein P and R1 are as defined above, followed by deprotection of the amine as described above, and coupling to
Rio Rll
L
wherein R10 and R11 are as *derfined above, W is a leaving group, such as mesylate, bromo or chloro, and L is leaving group such as halide, anhydride, active ester, and the like. For example when R10 and R11 are both hydrogen radical, bromoacetyl halide, chloroacetyl halide or the corresponding anhydride can be used. Finally, reacting the above intermediate with the cyclic-amine of formula
Figure imgf000046_0001
can produce the antiviral compounds of the present invention having the formula
Figure imgf000047_0001
wherein n, R1, R2, R3, R4, R10, R11, R12, R13 and R14 are as defined above. Amines of formula
Figure imgf000047_0002
are commercially available, such as pyrrolidine, 2- pyrrolidinemethanol, 3-pyrrolidinol, 2- (methoxymethyl) pyrrolidine, kainic acid, piperidine, 3- hydroxypiperidine, 4-hydroxypiperidine, 2- piperidinemethanol, 2,3-dihydroindole, isoindoline, 1,2,3, 4-tetrahydroisoquinoline, 1,2,3,4- tetrahydroquinoline, and the like; or can readily be prepared from commercially available starting materials using standard methods well known in the art, such as 4- hydroxyproline, 3-hydroxypiperidine, 4- hydroxypiperidine, 3-pyrroline, isonipecotic acid, 5- hydroxyindole, 5-hydroxyindole-3-acetic acid, 5,6- dimethoxyindole, isoquinoline, quinoline, 5- hydroxyisoquinoline, 8-hydroxy-5-nitroquinoline, 6,7- dimethoxy-1, 2, 3 ,4-tetrahydroisoquinoline, and the like. Alternatively, following neutralization of the salt, the amine is then coupled to the DL-, D-, or L- amino acid corresponding to the formula PNHCH(R1) COOH, wherein P and R1 are as defined above, followed by deprotection of the amine as described above and coupling the deprotected amine to the cyclic-amino acid of formula
Figure imgf000048_0001
wherein n, R10, R11, R12, R13 and R14 are as defined above, such as pyrrolidin-1-ylacetic acid, piperidin-1- ylacetic acid and the like, to produce the antiviral compounds of the present invention. The cyclic-amino acids are commercially available or are readily prepared from a protected carboxylic acid with a leaving group W (defined above) by reaction with the cyclic-amine as shown in Scheme III or alternatively, by reacting the cyclic-amine with the appropriate substituted ketone in the presence of cyanide anion followed by hydroylsis of the cyano group to the corresponding carboxylic acid as shown in Scheme IV, wherein n, P ,
Figure imgf000048_0002
R 14 are as defined above.
Scheme III
Figure imgf000048_0003
Scheme IV
Figure imgf000049_0001
Alternatively, following neutralization of the salt, the amine is then coupled to the DL-, D-, or L- amino acid corresponding to the formula
Figure imgf000049_0002
wherein n, R1, R 10 R 11 R12 R13 and R14 are as defined above, which can be prepared in a similar fashion to the coupling methods described above from DL-, D-, or L- amino acid corresponding to the formula NH CH(R )C00P3, wherein P3 and R1 are as defined above.
The DL-, D-, or L-amino acid corresponding to the formula PNHCH(R1) COOH or NH2CH(R1) COOP3 , wherein P, P3 and R are as defined above, are commercially available (Sigma Chemical Co.), or readily prepared using standard methods well known in the art from readily available starting materials. Preferably, P is a benzyloxycarbonyl or t-butoxycarbonyl radical and P3 is benzyl or tert-butyl radicals. Standard coupling procedures can be used to couple the amino acids and amines. The carboxylic acid group is reacted to form an anhydride, mixed anhydride, acid halide, such as chloride or bromide, or active ester, such as esters of N-hydroxysuccinimide, HOBT and the like, using well known procedures and conditions. Appropriate solvent systems include tetrahydrofuran, ethylether, methyl- tert-butylether, methylene chloride, N,N- dimethylformamide and the like, including mixtures thereof .
Alternatively, the protected amino alcohol from the epoxide opening can be further protected at the newly introduced amino group with a protecting group P" which is not removed with the removal of the amino protecting groups P or P1 and P2. One skilled in the art can choose appropriate combinations of P1, P, P1 and P2. For example, suitable combinations are P = Cbz and P' = Boc; P1 = Cbz and P = Bσc; P1 = Cbz, P2 = benzyl and P' = Boc; and P1 = P2 = benzyl and P1 = Boc. The resulting compound represented by the formula
R2 R2
Figure imgf000050_0001
can be carried through the remainder of the synthesis to provide a compound of the formula
Figure imgf000051_0001
wherein n, P', R1, R2 , R3, R10, R11, R12 , R13 and R14 are as defined above. The protecting group P' is then selectively removed and the resulting amine is reacted with the sulfonyl chloride RS02C1, the ,sulfonyl bromide R S02Br or the corresponding sulfonyl anhydride, preferably in the presence of an acid scavenger, to form the compounds of the present invention. This selective deprotection and conversion to the sulfonamide can be accomplished at either the end of the synthesis or at any appropriate intermediate step if desired.
The chemical reactions described above are generally disclosed in terms of their broadest application to the preparation of the compounds of this invention. Occasionally, the reactions may not be applicable as described to each compound included within the disclosed scope. The compounds for which this occurs will be readily recognized by those skilled in the art. In all such cases, either the reactions can be successfully performed by conventional modifications known to those skilled in the art, e.g., by appropriate protection of interfering groups, by changing to alternative conventional reagents, by routine modification of reaction conditions, and the like, or other reactions disclosed herein or otherwise conventional, will be applicable to the preparation of the corresponding compounds of this invention. In all preparative methods, all starting materials are known or readily prepared from known starting materials. Without further elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent. The following preferred specific embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever.
All reagents were used as received without purification. All proton and carbon NMR spectra were obtained on either a Varian VXR-300 or VXR-400 nuclear magnetic resonance spectrometer.
The following Examples illustrate the preparation of inhibitor compounds of the present invention and intermediates useful in preparing the inhibitor compounds of the present invention.
EXAMPLE 1
Figure imgf000052_0001
Preparation of 2S-fBis (phenylmethyl) aminoIbenzenepropanol
METHOD 1: 2S-[Bis (phenylmethyl) amino]benzenepropanol from the DIBAL Reduction of N,N-bis (phenylmethyl) -L- Phenylalanine phenylmethyl ester Step 1:
A solution of L-phenylalanine (50.0 g, 0.302 mol) , sodium hydroxide (24.2 g, 0.605 mol) and potassium carbonate (83.6 g, 0.605 mol) in water (500 mL) was heated to 97°C. Benzyl bromide (108.5 mL, 0.605 mol) was then slowly added (addition time -25 min) . The mixture was stirred at 97 °C for 30 minutes under a nitrogen atmosphere. The solution was cooled to room temperature and extracted with toluene (2 x 250 mL) . The combined organic layers were washed ,with water and brine, dried over magnesium sulfate, filtered and concentrated to an oil. The identity of the product was confirmed as follows. Analytical TLC (10% ethyl acetate/hexane, silica gel) showed major component at Rf value = 0.32 to be the "desired tribenzylated compound, N,N-bis (phenylmethyl) -L-phenylalanine phenylmethyl ester. This compound can be purified by column chromatography (silica gel, 15% ethyl acetate/hexane) . Usually the product is pure enough to be used directly in the next step without further purification. XH NMR spectrum was in agreement with published literature. XH NMR (CDCL3) d , 3.00 and 3.14 (ABX-system, 2H, JAB=14.1 Hz, AX=7.3 HZ and JBχ= 5.9 Hz), 3.54 and 3.92 (AB-System , 4 H, JAB=13.9 Hz), 3.71 (t, 1H, J=7.6 Hz), 5.11 and 5.23 (AB-System, 2H,
JAB=12.3 Hz), and 7.18 (m, 20 H) . EIMS: m/z 434 (M-l) .
Step 2:
The benzylated phenylalanine phenylmethyl ester (0.302 mol) from the previous reaction was dissolved in toluene (750 mL) and cooled to -55°C. A 1.5 M solution of DIBAL in toluene (443.9 mL, 0.666 mol) was added at a rate to maintain the temperature between -55 to -50°C (addition time -1 hr) . The mixture was stirred for 20 minutes under a nitrogen atmosphere and then quenched at -55°C by the slow addition of methanol (37 ml) . The cold solution was then poured into cold (5°C) 1.5 N HC1 solution (1.8 L) . The precipitated solid (approx. 138 g) was filtered off and washed with toluene. The solid material was suspended in a mixture of toluene (400 mL) and water (100 ml) . The mixture was cooled to 5°C and treated with 2.5 N NaOH (186 mL) and then stirred at room temperature until solid dissolved. The toluene layer was separated from the aqueous phase and washed with water and brine, dried over magnesium sulfate, filtered and concentrated to a volume of 75 mL (89 g) . Ethyl acetate (25 mL) and hexane (25 mL) were added to the residue upon which the desired alcohol product began to crystallize. After 30 min, an additional 50 mL hexane were added to promote further crystallization. The solid was filtered off and washed with 50 mL hexane to give 34.9 g of first crop product. A second crop of product (5.6 g) was isolated by refiltering the mother liquor. The two crops were combined and recrystallized from ethyl acetate (20 mL) and hexane (30 mL) to give 40 g of BS-2-[Bis (phenylmethyl) amino]benzenepropanol, 40% yield from L- phenylalanine. An additional 7 g (7%) of product can be obtained from recrystallization of the concentrated mother liquor. TLC of product Rf = 0.23 (10% ethyl acetate/hexane, silica gel) ^H NMR (CDC13) d 2.44 (m, 1H,), 3.09 (m, 2H) , 3.33 (m, 1H) , 3.48 and 3.92 (AB- System, 4H, JAB= 13.3 Hz), 3.52 (m, 1H) and 7.23 (m, 15H) ; [α]D25 +42.4 (c 1.45, CH2C12) ; DSC 77.67°C; Anal. Calcd. for C23H25ON: C, 83.34; H, 7.60; N, 4.23. Found: C, 83.43; H, 7.59; N, 4.22. HPLC on chiral stationary phase: Cyclobond I SP column (250 x 4.6 mm I.D.), mobile phase: methanol/triethyl ammonium acetate buffer pH 4.2 (58:42, v/v) , flow-rate of 0.5 ml/min, detection with detector at 230nm and a temperature of 0°C. Retention time: 11.25 min. , retention time of the desired product enantiomer: 12.5 min.
METHOD 2: Preparation of BS-2-[Bis (phenylmethyl) amino] benzene-propanol from the N,N-Dibenzylation of L- Phenylalaninol
L-phenylalaninol (176.6 g, 1.168 mol) was added to a stirred solution of potassium carbonate (484.6 g, 3.506 mol) in 710 mL of water. The mixture was heated to 65°C under a nitrogen atmosphere. A solution of benzyl bromide (400 g, 2.339 mol) in 3A ethanol (305 mL) was added at a rate that maintained the temperature between 60-68°C. The biphasic solution was stirred at 65°C for 55 min and then allowed to cool to 10°C with vigorous stirring. The oily product solidified into small granules. The product was diluted with 2.0 L of tap water and stirred for 5 minutes to dissolve the inorganic by products. The product was isolated by filtration under reduced pressure and washed with water until the pH is 7. The crude product obtained was air dried overnight to give a semi-dry solid (407 g) which was recrystallized from 1.1 L of ethyl acetate/heptane (1:10 by volume). The product was isolated by filtration (at -8°C) , washed with 1.6 L of cold (-10°C) ethyl acetate/heptane (1:10 by volume) and air-dried to give 339 g (88% yield) of BS-2- [Bis (phenylmethyl) amino]benzene-propanol, Mp = 71.5- 73.0°C. More product can be obtained from the mother liquor if necessary. The other analytical characterization was identical to compound prepared as described in Method 1. EXAMPLE 2
Figure imgf000056_0001
Preparation of 2s-[Bis (phenylmethyl) amino] benzenepropanaldehyde
METHOD 1: 2S- [Bis (phenylmethyl) amino]benzene-propanol (200 g, 0.604 mol) was dissolved in triethylamine (300 mL, 2.15 mol). The mixture was cooled to 12 °C and a solution of sulfur trioxide/pyridine complex (380 g, 2.39 mol) in DMSO (1.6 L) was added at a rate to maintain the temperature between 8-17°C (addition time -1.0 h) . The solution was stirred at ambient temperature under a nitrogen atmosphere for 1.5 hour at which time the reaction was complete by TLC analysis (33% ethyl acetate/hexane, silica gel) . The reaction mixture was cooled with ice water and quenched with 1.6 L of cold water (10-15°C) over 45 minutes. The resultant solution was extracted with ethyl acetate
(2.0 L) , washed with 5% citric acid (2.0 L) , and brine (2.2 L) , dried over MgS04 (280 g) and filtered. The solvent was removed on a rotary evaporator at 35-40°C and then dried under vacuum to give 198.8 g of 2s-[Bis- (phenylmethyl) amino] -benzenepropanaldehyde as a pale yellow oil (99.9%) . The crude product obtained was pure enough to be used directly in the next step without purification. The analytical data of the compound were consistent with the published literature. [α]D25 = -92.9°C (c 1.87, CH2C12) ; 1H NMR (400 MHz, CDC13) 3, 2.94 and 3.15 (ABX-System, 2H, JAB= 13.9 Hz, JM= 7.3 Hz and JBX = 6.2 Hz), 3.56 (t, 1H, 7.1 Hz), 3.69 and 3.82 (AB-System, 4H, JAB= 13.7 Hz), 7.25 (m, 15 H) and 9.72 (s, 1H) ; HRMS Calcd for (M+l) C23H24NO 330.450, found: 330.1836. Anal. Calcd. for
C23H23ON: C, 83.86; H, 7.04; N, 4.25. Found: C, 83.64; H, 7.42; N, 4.19. HPLC on chiral stationary phase: (S,S) Pirkle-Whelk-O 1 column (250 x 4.6 mm I.D.), mobile phase: hexane/isopropanol (99.5:0.5, v/v), flow-rate: 1.5 ml/min, detection with UV detector at 210nm. Retention time of the desired S-isomer: 8.75 min., retention time of the R-enantiomer 10.62 min.
METHOD 2: A solution of oxalyl chloride (8.4 ml, 0.096 mol) in dichloromethane (240 ml) was cooled to -74 °C. A solution of DMSO (12.0 ml, 0.155 mol) in dichloromethane (50 ml) was then slowly added at a rate to maintain the temperature at -74 °C (addition time "1.25 hr) . The mixture was stirred for 5 min. followed by addition of a solution of BS-2-
[bis (phenylmethyl) amino]benzene-propanol (0.074 mol) in 100 ml of dichloromethane (addition time -20 min. , temp. -75 °C to -68 °C) . The solution was stirred at -78 °C for 35 minutes under a nitrogen atmosphere. Triethylamine (41.2 ml, 0.295 mol) was then added over 10 min. (temp. -78° to -68°C) upon which the ammonium salt precipitated. The cold mixture was stirred for 30 min. and then water (225 ml) was added. The dichloromethane layer was separated from the aqueous phase and washed with water, brine, dried over magnesium sulfate, filtered and concentrated. The residue was diluted with ethyl acetate and hexane and then filtered to further remove the ammonium salt. The filtrate was concentrated to give αS- [bis (phenylmethyl) amino] benzenepropanaldehyde. The aldehyde was carried on to the next step without purification.
METHOD 3: To a mixture of 1.0 g(3.0 mmoles) of BS-2- [bis (phenylmethyl) amino] benzenepropanol 0.531 g(4.53 mmoles) of N-methyl morpholine, 2.27 g of molecular sieves (4A) and 9.1 mL of acetonitrile was added 53 mg (0.15 mmoles) of tetrapropylammonium perruthenate(TPAP) . The mixture was stirred for 40 minutes at room temperature and concentrated under reduced pressure. The residue was suspended in 15 mL of ethyl acetate, filtered through a pad of silica gel. The filtrate was concentrated under reduced pressure to give a product containing approximately 50% of αS-2- [bis (phenylmethyl) amino] benzene propanaldehyde as a pale yellow oil.
METHOD 4: To a solution of 1.0 g (3.02 mmoles) of BS- 2- [bis (phenylmethyl) amino]benzenepropanol in 9.0 mL of toluene was added 4.69 mg(0.03 mmoles) of 2,2,6,6- tetramethyl-1-piperidinyloxy, free radical (TEMPO) , 0.32g(3.11 mmoles) of sodium bromide, 9.0 mL of ethyl acetate and 1.5 mL of water. The mixture was cooled to 0 2C and an aqueous solution of 2.87 mL of 5% household bleach containing 0.735 g(8.75 mmoles) of sodium bicarbonate and 8.53 mL of water was added slowly over 25 minutes. The mixture was stirred at OSC for 60 minutes. Two more additions (1.44 mL each) of bleach was added followed by stirring for 10 minutes. The two phase mixture was allowed to separate. The aqueous layer was extracted twice with 20 mL of ethyl acetate. The combined organic layer was washed with 4.0 mL of a solution containing 25 mg of potassium iodide and water (4.0 mL) , 20 mL of 10% aqueous sodium thiosulfate solution and then brine solution. The organic solution was dried over magnesium sulfate, filtered and concentrated under reduced pressure to give 1.34g of crude oil containing a small amount of the desired product aldehyde, αS- [bis (phenylmethyl) amino] benzenepropanaldehyde.
METHOD 5: Following the same procedures as described in Method 1 of this Example except 3.0 equivalents of sulfur trioxide pyridine complex was used and αS- [bis (phenylmethyl) amino]benzenepropanaldehyde was isolated in comparable yields.
EXAMPLE 3
Figure imgf000060_0001
Preparation of N,N-dibenzyl-3 (S) -amino-1, 2-(S) -epoxy-4-phenylbutane
METHOD 1: A solution of αS-
[Bis (phenylmethyl) amino]benzene-propanaldehyde (191.7g,
0.58 mol) and chloroiodomethane (56.4 mL, 0.77 mol) in tetrahydrofuran (1.8 L) was cooled to -30 to -35°C
(colder temperature such as -70°C also worked well but warmer temperatures are more readily achieved in large scale operations) in a stainless steel reactor under a nitrogen atmosphere. A solution of n-butyl lithium in hexane (1.6 M, 365 mL, 0.58 mol) was then added at a rate that maintained the temperature below -25°C. After addition the mixture was stirred at -30 to -35°C for 10 minutes. More additions of reagents were carried out in the following manner: (1) additional chloroiodomethane (17 mL) was added, followed by n-butyl lithium (110 mL) at <-25°C. After addition the mixture was stirred at -30 to -35°C for 10 minutes. This was repeated once.
(2) Additional chloroiodomethane (8.5 mL, 0.11 mol) was added, followed by n-butyl lithium (55 mL, 0.088 mol) at <-25°C. After addition the mixture was stirred at -30 to -35°C for 10 minutes. This was repeated 5 times.
(3) Additional chloroiodomethane (8.5 mL, 0.11 mol) was added, followed by n-butyl lithium (37 mL, 0.059 mol) at <-25°C. After addition the mixture was stirred at -30 to -35°C for 10 minutes. This was repeated once. The external cooling was stopped and the mixture warmed to ambient temp, over 4 to 16 hours when TLC (silica gel, 20% ethyl acetate/hexane) indicated that the reaction was completed. The reaction mixture was cooled to 10°C and quenched with 1452g of 16% ammonium chloride solution (prepared by dissolving 232g of ammonium chloride in 1220 mL of water) , keeping the temperature below 23 °C. The mixture was stirred for 10 minutes and the organic and aqueous layers were separated. The aqueous phase was extracted with ethyl acetate (2x 500 mL) . The ethyl acetate layer was combined with the tetrahydrofuran layer. The combined solution was dried over magnesium sulfate (220g) , filtered and concentrated on a rotary evaporator at 65°C. The brown oil residue was dried at 70°C in vacuo (0.8 bar) for 1 h to give 222.8g of crude material. (The crude product weight was >100%. Due to the relative instability of the product on silica gel, the crude product is usually used directly in the next step without purification) . The diastereomeric ratio of the crude mixture was determined by proton NMR: (2S)/(2R): 86:14. The minor and major epoxide diastereomers were characterized in this mixture by tic analysis (silica gel, 10% ethyl acetate/hexane) , Rf = 0.29 & 0.32, respectively. An analytical sample of each of the diastereomers was obtained by purification on silica-gel chromatography (3% ethyl acetate/hexane) and characterized as follows:
N,N,αS-Tris (phenylmethyl) -2S-oxiranemethanamine
1H NMR (400 MHz, CDCl3) d 2.49 and 2.51 (AB-System, 1H, JAB = 2.82), 2.76 and 2.77 (AB-System, 1H, JAB = 4.03), 2.83 (m, 2H) , 2.99 & 3.03 (AB-System, 1H, JAB = 10.1 Hz) , 3.15 (m, 1H) , 3.73 & 3.84 (AB-System, 4H, JAB = 14.00), 7.21 (m, 15H) ; 13C NMR (400 MHz,CDCl3) 8 139.55, 129.45, 128.42, 128.14, 128.09, 126.84, 125.97, 60.32, 54.23, 52.13, 45.99, 33.76; HRMS Calcd for C24H26NO (M+l) 344.477, found 344.2003.
N,N,αS-Tris (phenylmethyl) -2R-oxiranemethanamine
1H NMR (300 MHz, CDC13) d 2.20 (m, 1H) , 2.59 (m, 1H) , 2.75 (m, 2H) , 2.97 (m, 1H) , 3.14 (m, 1H) , 3.85 (AB- System, 4H) , 7.25 (m, 15H) .HPLC on chiral stationary phase: Pirkle-Whelk-0 1 column (250 x 4.6 mm I.D.), mobile phase: hexane/isopropanol (99.5:0.5, v/v), flow- rate: 1.5 ml/min, detection with UV detector at 210nm. Retention time of (8) : 9.38 min., retention time of enantiomer of (4): 13.75 min.
METHOD 2: A solution of the crude aldehyde 0.074 mol and chloroiodomethane (7.0 ml, 0.096 mol) in tetrahydrofuran (285 ml) was cooled to -78°C, under a nitrogen atmosphere. A 1.6 M solution of n-butyl lithium in hexane (25 ml, 0.040 mol) was then added at a rate to maintain the temperature at -75°C (addition time - 15 min.). After the first addition, additional chloroiodomethane (1.6 ml, 0.022 mol) was added again, followed by n-butyl lithium (23 ml, 0.037 mol), keeping the temperature at -75°C. The mixture was stirred for 15 min. Each of the reagents, chloroiodomethane (0.70 ml, 0.010 mol) and ή-butyl lithium (5 ml, 0.008 mol) were added 4 more times over 45 min. at -75°C. The cooling bath was then removed and the solution warmed to 22 °C over 1.5 hr. The mixture was poured into 300 ml of saturated aq. ammonium chloride solution. The tetrahydrofuran layer was separated. The aqueous phase was extracted with ethyl acetate (1 x 300 ml) . The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated to give a brown oil (27.4g). The product could be used in the next step without purification. The desired diastereomer can be purified by recrystallization at a subsequent step. The product could also be purified by chromatography.
METHOD 3: A solution of αS-
[Bis (phenylmethyl) amino]benzene-propanaldehyde (178.84g, 0.54 mol) and bromochloromethane (46 mL, 0.71 mol) in tetrahydrofuran (1.8 L) was cooled to -30 to -35°C (colder temperature such as -70°C also worked well but warmer temperatures are more readily achieved in large scale operations) in a stainless steel reactor under a nitrogen atmosphere. A solution of n-butyl lithium in hexane (1.6 M, 340 mL, 0.54 mol) was then added at a rate that maintained the temperature below -25°C. After addition the mixture was stirred at -30 to -35°C for 10 minutes. More additions of reagents were carried out in the following manner: (1) additional bromochloromethane (14 mL) was added, followed by n-butyl lithium (102 mL) at < -25°C. After addition the mixture was stirred at -30 to -35°C for 10 minutes. This was repeated once. (2) Additional bromochloromethane (7 mL, 0.11 mol) was added, followed by n-butyl lithium (51 mL, 0.082 mol) at <-25°C. After addition the mixture was stirred at -30 to -35°C for 10 minutes. This was repeated 5 times. (3) Additional bromochloromethane (7 mL, 0.11 mol) was added, followed by n-butyl lithium (51 mL, 0.082 mol) at <-25°C. After addition the mixture was stirred at -30 to -35°C for 10 minutes. This was repeated once. The external cooling was stopped and the mixture warmed to ambient temp, over 4 to 16 hours when TLC (silica gel, 20% ethyl acetate/hexane) indicated that the reaction was completed. The reaction mixture was cooled to 10°C and quenched with 1452g of 16% ammonium chloride solution (prepared by dissolving 232g of ammonium chloride in 1220 mL of water) , keeping the temperature below 23 °C. The mixture was stirred for 10 minutes and the organic and aqueous layers were separated. The aqueous phase was extracted with ethyl acetate (2x 500 mL) . The ethyl acetate layer was combined with the tetrahydrofuran layer. The combined solution was dried over magnesium sulfate (220g) , filtered and concentrated on a rotary evaporator at 65°C. The brown oil residue was dried at 70°C in vacuo (0.8 bar) for 1 h to give 222.8g of crude material.
METHOD 4: Following the same procedures as described in Method 3 of this Example except the reaction temperatures were at -20°C. The resulting N,N,αS- tris (phenylmethyl) -2S-oxiranemethanamine was a diastereomeric mixture of lesser purity then that of Method 3.
METHOD 5: Following the same procedures as described in Method 3 of this Example except the reaction temperatures were at -70 to -78°C. The resulting N,N,αS-tris (phenylmethyl) -2S-oxiranemethanamine was a diastereomeric mixture, which was used directly in the subsequent steps without purification.
METHOD 6: Following the same procedures as described in Method 3 of this Example except a continuous addition of bromochloromethane and n-butyl lithium was used at -30 to -35°C. After the reaction and work up procedures as described in Method 3 of this Example, the desired N,N,αS-tris (phenylmethyl) -2S-oxiranemethanamine was isolated in comparable yields and purities.
METHOD 7: Following the same procedures as described in Method 2 of this Example except dibromomethane was used instead of chloroiodomethane. After the reaction and work up procedures as described in Method 2 of this Example, the desired N,N, S-tris (phenylmethyl) -2S- oxirane-methanamine was isolated.
EXAMPLE 4
Figure imgf000065_0001
Preparation of N- f3 (S) -[N,N-bis (phenylmethyl) amino] - 2 (R) -hydroxy-4-phenylbutyl]-N-isobutylamine
To a solution of crude N,N-dibenzyl-3 (S) -amino- 1,2 (S)-epoxy-4-phenylbutane (388.5g, 1.13 mol) in isopropanol (2.7 L) (or ethyl acetate) was added isobutylamine (1.7 kgm, 23.1 mol) over 2 min. The temperature increased from 25°C and to 30°C. The solution was heated to 82 °C and stirred at this temperature for 1.5 hours. The warm solution was concentrated under reduced pressure at 65°C, The brown oil residue was transferred to a 3-L flask and dried in vacuo (0.8 mm Hg) for 16 h to give 450g of 3S-[N,N- bis (phenylmethyl) amino-4-phenylbutan-2R-ol as a crude oil.
An analytical sample of the desired major diastereomeric product was obtained by purifying a small sample of crude product by silica gel chromatography
(40% ethyl acetate/hexane) . Tic analysis: silica gel, 40% ethyl acetate/hexane; Rf = 0.28; HPLC analysis: ultrasphere ODS column, 25% triethylamino-/phosphate buffer pH 3-acetonitrile, flow rate 1 mL/min, UV detector; retention time 7.49 min.; HRMS Calcd for C28H27N20 (M + 1) 417.616, found 417.2887. An analytical sample of the minor diastereomeric product, 3S-[N,N- bis (phenylmethyl) amino] 1- (2-methylpropyl) amino-4- phenylbutan-2S-ol was also obtained by purifying a small sample of crude product by silica gel chromatography (40% ethyl acetate/hexane) .
EXAMPLE 5
Figure imgf000066_0001
Preparation of N- r 3 (S)-[N,N-bis (phenylmethyl) amino] -2 (R) -hydroxy-4-phenylbutyl]-N- isobutylamine'oxalic acid salt
To a solution of oxalic acid (8.08g, 89.72 mmol) in methanol (76 mL) was added a solution of crude 3(S)- [N,N-bis (phenylmeth 1) amino]-l-(2-methylpropyl) amino-4- phenylbutan-2 (R) -ol {39.68g, which contains about 25.44g (61.06 mmol) of 3(S),2(R) isomer and about 4.49g
(10.78 mmol) of 3(S),2(S) isomer} in ethyl acetate (90 mL) over 15 minutes. The mixture was stirred at room temperature for about 2 hours. Solid was isolated by filtration, washed with ethyl acetate (2 x 20 mL) and dried in vacuo for about 1 hour to yield 21.86g (70.7% isomer recovery) of 97% diastereomerically pure salt (based on HPLC peak areas) . HPLC analysis: Vydec- peptide/protein C18 column, UV detector 254 nm, flow rate 2 mL/min. , gradient {A = 0.05% trifluoroacetic acid in water, B = 0.05% trifluoroacetic acid in acetonitrile, 0 min. 75% A/25% B, 30 min. 10% A/90% B, 35 min. 10% A/90% B, 37 min. 75% A/25% B}; Retention time 10.68 min. (3(S),2(R) isomer) and 9.73 min. (3(S),2(S) isomer). Mp = 174.99°C; Microanalysis: Calc: C 71.05%, H 7.50%, -N 5.53%; Found: C 71.71%, H 7.75%, N 5.39%.
Alternatively, oxalic acid dihydrate (119g, 0.94 mole) was added to a 5000 mL round bottom flask fitted with a mechanical stirrer and a dropping funnel. Methanol (1000 ml) was added and the mixture stirred until dissolution was complete. A solution of crude 3 (S) - [N,N-bis (phenylmethyl) amino] -1- (2-methylpropyl) amino-4-phenylbutan-2 (R) -ol in ethyl acetate (1800 ml, 0.212g amino alcohol isomers/mL, 0.9160 moles) was added over a twenty minute period. The mixture was stirred for 18 hours and the solid product was isolated by centrifugation in six portions at 400G. Each portion was washed with 125 mL of ethyl acetate. The salt was then collected and dried overnight at 1 torr to yield 336.3g of product (71% based upon total amino alcohol). HPLC/MS (electrospray) was consistent with the desired product (m/z 417 [M+H]+) .
Alternatively, crude 3 (S) -[N,N-bis (phenylmethyl) amino] -1- (2-methylpropyl) amino-4-phenylbutan-2 (R) -ol (5g) was dissolved in ethyl-tert-butylether (MTBE) (10 mL) and oxalic acid (lg) in methanol (4 mL) was added. The mixture was stirred for about 2 hours. The resulting solid was filtered, washed with cold MTBE and dried to yield 2. lg of white solid of about 98.9% diastereomerically pure (based on HPLC peak areas) . EXAMPLE 6
Preparation of N-[3 (S) -fN,N-bis (phenylmethyl) amino] -2 (R) -hydroχy-4-phenylbutyll-N- isobutylamine»acetic acid salt
To a solution of crude 3 (S) -[N,N-bis (phenylmethyl) amino] -l-(2-methylpropyl) amino-4-phenylbutan-2 (R)-ol in methyl-tert-butylether (MTBE) (45 mL, l.lg amino alcohol isomers/mL) was added acetic acid (6.9 mL) dropwise. The mixture was stirred for about 1 hour at room temperature. The solvent was removed in vacuo to yield a brown oil about 85% diastereomerically pure product (based on HPLC peak areas) . The brown oil was crystallized as follows: 0.2g of the oil was dissolved in the first solvent with heat to obtain a clear solution, the second solvent was added until the solution became cloudy, the mixture was heated again to clarity, seeded with about 99% diastereomerically pure product, cooled to room temperature and then stored in a refrigerator overnight. The crystals were filtered, washed with the second solvent and dried. The diastereomeric purity of the crystals was calculated from the HPLC peak areas. The results are shown in Table 1.
TABLE 1
Figure imgf000068_0001
Alternatively, crude 3 (S) -[N, N-bis (phenylmethyl) amino] -1- (2-methylpropyl) amino-4-phenylbutan-2 (R) -ol {50. Og, which contains about 30.06g (76.95 mmol) of 3(S),2(R) isomer and about 5.66g (13.58 mmol) of 3(S),2(S) isomer} was dissolved in methyl-tert- butylether (45.0 mL) . To this solution was added acetic acid (6.90 mL, 120.6 mmol) over a period of about 10 min. The mixture was stirred at room temperature for about 1 hour and concentrated under reduced pressure. The oily residue was purified by recrystallization from methyl-tert-butylether (32 mL) and heptane (320 L) . Solid was isolated by filtration, washed with cold heppane and dried in vacuo for about 1 hour to afford 21.34c (58.2% isomer recovery) of 96% diastereomerically pure monoacetic acid salt (based on HPLC peak areas) . Mp = 105-106°C; Microanalysis: Calc. : C 75.53%, H 8.39%, N 5.87%; Found: C 75.05%, H 8.75%, N 5.71%.
EXAMPLE 7
Preparation of N-[3 (s) - N.-N-bis (phenylmethyl) amino] -2 (R) -hvdroxy-4-phenylbutyl]-N- isobutylamine»L-tartaric acid salt
Crude 3 (S)-[N, N-bis (phenylmethyl) amino] -1- (2- methylpropyl) amino-4-phenylbutan-2 (R) -ol {10.48g, which contains about 6.72g (16.13 mmol) of 3(S),2(R) isomer and about 1.19g (2.85 mmol) of 3(S),2(S) isomer} was dissolved in tetrahydrofuran (10.0 mL) . To this solution was added a solution of L-tartaric acid (2.85g, 19 mmol) in methanol (5.0 mL) over a period of about 5 min. The mixture was stirred at room temperature for about 10 min. and concentrated under reduced pressure. Methyl-tert-butylether (20.0 mL) was added to the oily residue and the mixture was stirred at room temperature for about 1 hour. Solid was isolated by filtration to afford 7.50g of crude salt. The crude salt was purified by recrystallization from ethyl acetate and heptane at room temperature to yield 4.13g (45.2% isomer recovery) of 95% diastereomerically pure L-tartaric acid salt (based on HPLC peak areas). Microanalysis: Calc. : C 67.76%, H 7.41%, N 4.94%; Found: C 70.06%, H 7.47%, N 5.07%.
EXAMPLE 8
Preparation of N-T3 (S) -fN;N-bis (phenylmάthyl) aminol 2 (R) -hydroxy-4-phenylbutyl]-N- isobutylamine'dihvdrochloric acid salt
Crude 3 (S)-[N, N-bis (phenylmethyl) amino] -1- (2- methylpropyl) amino-4-phenylbutan-2 (R) -ol {10. Og, which contains about 6.41g (15.39 mmol) of 3(S),2(R) isomer and about 1.13g (2.72 mmol) of 3(S),2(S) isomer} was dissolved in tetrahydrofuran (20.0 mL) . To this solution was added hydrochloric acid (20 mL, 6.0 N) over a period of about 5 min. The mixture was stirred at room temperature for about 1 hour and concentrated under reduced pressure. The residue was recrystallized from ethanol at 0°C to yield 3.20g (42.7% isomer recovery) of 98% diastereomerically pure dihydrochloric acid salt (based on HPLC peak areas). Microanalysis: Calc: C 68.64%, H 7.76%, N 5.72%; Found: C 68.79%, H 8.07%, N 5.55%. EXAMPLE 9
Preparation of N-r 3 (S) -fN,N-bis (phenylmethyl) amino] - 2 (R) -hydroxy-4-phenylbuty1] -N- isobutylamine'toluenesulfonic acid salt
Crude 3 (S) -[N,N-bis (phenylmethyl) amino] -1- (2- methylpropyl) amino-4-phenylbutan-2 (R) -ol {5.0g, which contains about 3.18g (7.63 mmol) of 3(S),2(R) isomer and about 0.56g (1.35 mmol) of 3(S),2(S) isomer} was dissolved in methyl-tert-butylether (10*.0 mL) . To this solution was added a solution of toluenesulfonic acid (2.28g, 12 mmol) in methyl-tert-butylether (2.0 mL) and methanol (2.0 mL) over a period of about 5 min. The mixture was stirred at room temperature for about 2 hours and concentrated under reduced pressure. The residue was recrystallized from methyl-tert-butylether and heptane at 0°C, filtered, washed with cold heptane and dried in vacuo to yield 1.85g (40.0% isomer recovery) of 97% diastereomerically pure monotoluenesulfonic acid salt (based on HTLC peak areas) .
EXAMPLE 10
Preparation of N-[ 3 (S) -TN,N-bis (phenylmethyl) aminoV 2 (R) -hydroxy-4-phenylbutyll-N- isobutylamine'methanesulfonic acid salt
Crude 3 (S) -[N,N-bis (phenylmethyl) amino]-l-(2- methylpropyl) amino-4-phenylbutan-2 (R) -ol {10.68g, which contains about 6.85g (16.44 mmol) of 3(S),2(R) isomer and about 1.21g (2.90 mmol) of 3(S),2(S) isomer} was dissolved in tetrahydrofuran (10.0 mL) . To this solution was added methanesulfonic acid (1.25 mL, 19.26 mmol). The mixture was stirred at room temperature for about 2 hours and concentrated under reduced pressure. The oily residue was recrystallized from methanol and water at 0°C, filtered, washed with cold methanol/water (1:4) and dried in vacuo to yield 2.40g (28.5% isomer recovery) of 98% diastereomerically pure monomethanesulfonic acid salt (based on HPLC peak areas) .
EXAMPLE 11
Preparation of N-benzyl-L-phenylalaninol
METHOD 1: L-Phenylalaninol (89.51g, 0.592 moles) was dissolved in 375 mL of methanol under inert atmosphere, 35.52g (0.592 moles) of glacial acetic acid and 50 mL of methanol was added followed by a solution of 62.83g (0.592 moles) of benzaldehyde in 100 mL of methanol. The mixture was cooled to approximately 15°C and a solution of 134.6g (2.14 moles) of sodium cyanoborohydride in 700 mL of methanol was added in approximately 40 minutes, keeping the temperature between 15°C and 25°C. The mixture was stirred at room temperature for 18 hours. The mixture was concentrated under reduced pressure and partitioned between 1 L of 2M ammonium hydroxide solution and 2 L of ether. The ether layer was washed with 1 L of 1M ammonium hydroxide solution, twice with 500 mL water, 500 mL of brine and dried over magnesium sulfate for 1 hour. The ether layer was filtered, concentrated under reduced pressure and the crude solid product was recrystallized from 110 mL of ethyl acetate and 1.3 L of hexane to give 115g (81% yield) of N-benzyl-L-phenylalaninol as a white solid.
METHOD 2: L-Phenylalaninol (5g, 33 mmoles) and 3.59g (33.83 mmoles) of benzaldehyde were dissolved in 55 mL of 3A ethanol under inert atmosphere in a Parr shaker and the mixture was warmed to 60°C for 2.7 hours. The mixture was cooled to approximately 25 °C and 0.99g of 5% platinum on carbon was added and the mixture was hydrogenated at 60 psi of hydrogen and 40 °C for 10 hours. The catalyst was filtered off, the product was concentrated under reduced pressure and the crude solid product was recrystallized from 150 mL of heptane to give 3.83g (48 % yield) of N-benzyl-L-phenylalaninol as a white solid.
EXAMPLE 12
Preparation of N-(t-Butoxycarbonyl) -N- benzyl-L-phenylalaninol
N-benzyl-L-phenylalaninol (2.9g, 12 mmoles) was dissolved in 3 mL of triethylamine and 27 mL of methanol and 5.25g (24.1 mmoles) of di-tert-butyl dicarbonate was added. The mixture was warmed to 60°C for 35 minutes and concentrated under reduced pressure. The residue was dissolved in 150 mL of ethyl acetate and washed twice with 10 mL of cold (0-5°C) , dilute hydrochloric acid (pH 2.5 to 3), 15 mL of water, 10 mL of brine, dried over magnesium sulfate, filtered and concentrated under reduced pressure. The crude product oil was purified by silica gel chromatography (ethyl acetate: hexane, 12:3 as eluting solvent) to give 3.98g (97% yield) of colorless oil.
EXAMPLE 13
Preparation of N- (t-Butoxycarbonyl) -N- benzyl-L-phenylalaninal
METHOD 1: To a solution of 0.32 g(0.94 mmoles) of N-(t- butoxycarbonyl) -N-benzyl-L-phenylalaninol in 2.8 mL of toluene was added 2.4 mg (0.015 mmoles) of 2,2,6,6- tetramethyl-1-piperidinyloxy, free radical (TEMPO), O.lg (0.97 mmoles) of sodium bromide, 2.8 mL of ethyl acetate and 0.34 mL of water. The mixture was cooled to osc and an aqueous solution of 4.2 mL of 5% household bleach containing 0.23g (3.0 mL, 2.738 mmoles) of sodium bicarbonate was added slowly over 30 minutes. The mixture was stirred at OSC for 10 minutes. Three more additions (0.4 mL each) of bleach was added followed by stirring for 10 minutes after each addition to consume all the starting material. The two phase mixture was allowed to separate. The aqueous layer was extracted twice with 8 mL of toluene. The combined organic layer was washed with 1.25 mL of a solution containing 0.075g of potassium iodide, sodium bisulfate(0.125 g) and water (1.1 mL) , 1.25 mL of 10% aqueous sodium thiosulfate solution, 1.25 mL of pH 7 phosphate buffer and 1.5 mL of brine solution. The organic solution was dried over magnesium sulfate, filtered and concentrated under reduced pressure to give 0.32g (100% yield) of N-(t- Butoxycarbonyl) -N-benzyl-L-phenylalaninal.
METHOD 2: To a solution of 2.38 g(6.98 mmoles) of N-(t- butoxycarbonyl) -N-benzyl-L-phenylalaninol in 3.8 mL (27.2 mmoles) of triethylamine at lO^C was added a solution of 4.33g (27.2 mmoles) of sulfur trioxide pyridine complex in 17 mL of dimethyl sulfoxide. The mixture was warmed to room temperature and stirred for one hour. Water (16 mL) was added and the mixture was extracted with 20 mL of ethyl acetate. The organic layer was washed with 20 mL of 5% citric acid, 20 mL of water, 20 L of brine, dried over magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure to give 2.37g (100% yield) of N-(t- Butoxycarbony1) -N-benzyl-L-phenylalaninal . EXAMPLE 14
Figure imgf000075_0001
Preparation of 3 (S) -[N- (t-butoxycarbonyl) -N- benzylamino]-1.2-(S) -epoxy-4-phenylbutane
METHOD 1: A solution of 2.5g (7.37 mmoles) of N-(t- butoxycarbonyl) -N-benzyl-L-phenylalaninal and 0.72 mL of chloroiodomethane in 35 mL of THF was cooled to -78 °C. A 4.64 mL of a solution of n-butyllithium (1.6 M in hexane, 7.42 mmoles) was added slowly, keeping the temperature below -70°C. The mixture was stirred for 10 minutes between -70 to -75°C. Two additional portions of 0.22 mL of chloroiodomethane and 1.4 mL of n- butyllithium was added sequentially and the mixture was stirred for 10 minutes between -70 to -752C after each addition. Four additional portions of 0.11 mL of chloroiodomethane and 0.7 mL of n-butyllithium was added sequentially and the mixture was stirred for 10 minutes between -70 to -75°C after each addition. The mixture was warmed to room temperature for 3.5 hours . The product was quenched at below 5°C with 24 mL of ice-cold water. The biphasic layers were separated and the aqueous layer was extracted twice with 30 mL of ethyl acetate. The combined organic layers was washed three times with 10 mL water, then with 10 mL brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 2.8g of a yellow crude oil. This crude oil (>100% yield) is a mixture of the diastereomeric epoxides N,αS-bis (phenylmethyl) -N- (t- butoxycarbonyl) -2S-oxiranemethanamine and N,αS- bis (phenylmethyl) -N- (t-butoxycarbonyl) -2R- oxiranemethanamine. The crude mixture is used directly in the next step without purification.
METHOD 2: To a suspension of 2.92g (13.28 mmoles) of trimethylsulfoxonium iodide in 45 mL of acetonitrile was added 1.49g (13.28 mmoles) of potassium t-butoxide. A solution of 3.0g (8.85 mmoles) of N- (t-butoxycarbonyl) - N-benzyl-L-phenylalaninal - in 18 mL of acetonitrile was added and the mixture was stirred at room temperature for one hour. The mixture was diluted with 150 mL of water and extracted twice with 200 mL of ethyl acetate. The organic layers were combined and washed with 100 mL water, 50 mL brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 3. Og of a yellow crude oil. The crude product was purified by silica gel chromatography (ethyl acetate/hexane: 1: 8 as eluting solvent) to give 1.02g (32.7% yield) of a mixture of the two diastereomers N,αS-bis (phenylmethyl) - N- (t-butoxycarbonyl) -2S-oxiranemethanamine and N,αS- bis (phenylmethyl) -N- (t-butoxycarbonyl) -2R- oxiranemethanamine.
METHOD 3: To a suspension of 0.90g (4.42 mmoles) of trimethylsulfonium iodide in 18 mL of acetonitrile was added 0.495g (4.42 mmoles) of potassium t-butoxide. A solution of l.Og (2.95 mmoles) of N- (t-butoxycarbonyl) - N-benzyl-L-phenylalaninal in 7 mL of acetonitrile was added and the mixture was stirred at room temperature for one hour. The mixture was diluted with 80 mL of water and extracted twice with 80 mL of ethyl acetate. The organic layers were combined and washed with 100 mL water, 30 mL brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1.04g of a yellow crude oil. The crude product was a mixture of the two diastereomers N,αS-bis (phenylmethyl) -N- (t- butoxycarbonyl) -2S-oxiranemethanamine and N,αS- bis (phenylmethyl) -N- (t-butoxycarbonyl) -2R- oxiranemethanamine.
EXAMPLE 15
Figure imgf000077_0001
Preparation of 3S-[N- (t-Butoxycarbonyl) -N-( phenylmethyl) amino]-!- (2-methylpropyl) amino-4-phenylbutan-2R-ol
To a solution of 500 mg (1.42 mmoles) of the crude epoxide (a mixture of the two diastereomers N,αS- bis (phenylmethyl) -N- (t-butoxycarbonyl) -2S- oxiranemethanamine and N,αS-bis (phenylmethyl) -N- (t- butoxycarbony1) -2R-oxiranemethanamine) in 0.98 mL of isopropanol was added 0.71 mL (7.14 mmoles) of isobutylamine. The mixture was warmed to reflux at 85°C to 90°C for 1.5 hours. The mixture was concentrated under reduced pressure and the product oil was purified by silica gel chromatography (chloroform:methanol, 100:6 as eluting solvents) to give 330 mg of 3S-[N-(t- butoxycarbonyl) -N- (phenylmethyl) amino] -1- (2- methylpropyl) amino-4-phenylbutan-2R-ol as a colorless oil (54.5% yield). 3S- [N- (t-Butoxycarbonyl) -N- (phenylmethy1) amino]-l- (2-methylpropyl) amino-4- phenylbutan-2S-ol was also isolated. When purified N,αS-bis (phenylmethyl) -N- (t-butoxycarbonyl) -2S- oxiranemethanamine was used as starting material, 3S-[N- (t-butoxycarbonyl) -N- (phenylmethyl) amino] -1- (2- methylpropyl) amino-4-phenylbutan-2R-ol was isolated after purification by chromatography in an 86% yield.
EXAMPLE 16
Figure imgf000078_0001
Preparation of 3S-(N-t-Butoxycarbonyl) amino-4- phenylbutan-1 , 2R-diol
To a solution of lg (3.39 mmoles) of 2S-(N-t- butoxycarbon 1) amino-IS-hydroxy-3-phenylbutanoic acid (commercially available from Nippon Kayaku, Japan) in 50 mL of THF at 02C was added 50 mL of borane-THF complex (liquid, 1.0 M in THF), keeping the temperatures below 5°C. The reaction mixture was warmed to room temperature and stirred for 16 hours. The mixture was cooled to 0°C and 20 mL of water was added slowly to destroy the excess BH3 and to quench the product mixture, keeping the temperature below 12 °C. The quenched mixture was stirred for 20 minutes and concentrated under reduced pressure. The product mixture was extracted three times with 60 mL of ethyl acetate. The organic layers were combined and washed with 20 mL of water, 25 mL of saturated sodium chloride solution and concentrated under reduced pressure to give l.lg of crude oil. The crude product was purified by silica gel chromatography (chloroform/methanol, 10:6 as eluting solvents) to give 900 mg (94.4% yield) of 3S-(N- t-butoxycarbonyl) amino-4-phenylbutan-l, 2R-diol as a white solid. EXAMPLE 17
Figure imgf000079_0001
Preparation of 3S-(N-t-Butoxycarbonyl) amino-2R-hydroxy- 4-phenylbut-l-yl Toluenesulfonate
To a solution of 744.8 mg (2.65 mmoles) of 3S-(N-t- butoxycarbonyl)amino-4-phenylbutan-l,2R-diol in 13 mL of pyridine at O^c was added 914 mg of toluenesulfonyl chloride in one portion. The mixture was stirred at 0°C to 5°C for 5 hours. A mixture of 6.5 mL of ethyl acetate and 15 mL of 5% aqueous sodium bicarbonate solution was added to the reaction mixture and stirred for 5 minutes. The product mixture was extracted three times with 50 mL of ethyl acetate. The organic layers were combined and washed with 15 mL of water, 10 mL of saturated sodium chloride solution and concentrated under reduced pressure to give about l.lg of a yellow chunky solid. The crude product was purified by silica gel chromatography (ethyl acetate/hexane 1:3 as eluting solvents) to give 850 mg (74% yield) of 3S-(N-t- butoxycarbonyl) amino-2R-hydroxy-4-phenylbut-l-yl toluenesulfonate as a white solid. EXAMPLE 18
Figure imgf000080_0001
Preparation of 3S-[N- (t-Butoxycarbonyl) amino]-!- (2- methylpropyl) amino-4-phenylbutan-2R-ol
To a solution of 90 mg (0.207 mmoles) of 3S-(N-t- butoxycarbonyl) amino-2R-hydroxy-4-phenylbut-l) yl toluanesulfonate in 4.143 mL of isopropanol and 0.5 iL of toluene was added 0.103 mL (1.034 mmoles) of isobutylamine. The mixture was warmed to 80 to 85°C and stirred for 1.5 hours. The product mixture was concentrated under reduced pressure at 40 to 50°C and purified by silica gel chromatography (chloroform/methanol, 10:1 as eluting solvents) to give 54.9 mg (76.8% yield) of 3S-[N- (t-butoxycarbonyl) amino] - 1- (2-methylpropyl) amino-4-phenylbutan-2R-ol as a white solid.
EXAMPLE 19
Figure imgf000081_0001
Preparation of N- r 3 (S) -benzyloxycarbonylamino-2 (R) - hydroxy-4-phenylbutyl1 -N-isoamylamine *
Part A: To a solution of 75. Og (0.226 mol) of N- benzyloxycarbonyl-L-phenylalanine chloromethyl ketone in a mixture of 807 mL of methanol and 807 mL of tetrahydrofuran at -2°C, was added 13.17g (0.348 mol, 1.54 equiv.) of solid sodium borohydride over one hundred minutes. The solvents were removed under reduced pressure at 40°C and the residue dissolved in ethyl acetate (approx. IL) . The solution was washed sequentially with 1M potassium hydrogen sulfate, saturated sodium bicarbonate and then saturated sodium chloride solutions. After drying over anhydrous magnesium sulfate and filtering, the solution was removed under reduced pressure. To the resulting oil was added hexane (approx. IL) and the mixture warmed to 60°C with swirling. After cooling to room temperature, the solids were collected and washed with 2L of hexane. The resulting solid was recrystallized from hot ethyl acetate and hexane to afford 32.3g (43% yield) of N- benzyloxycarbonyl-3 (S) -amino-l-chloro-4-phenyl-2 (S) - butanol, mp 150-151°C and M+Li+ = 340.
Part B: To a solution of 6.52g (0.116 mol, 1.2 equiv.) of potassium hydroxide in 968 L of absolute ethanol at room temperature, was added 32.3g (0.097 mol) of N-CBZ- 3 (S) -amino-l-chloro-4-phenyl-2 (S) -butanol. After stirring for fifteen minutes, the solvent was removed under reduced pressure and the solids dissolved in methylene chloride. After washing with water, drying over magnesium sulfate, filtering and stripping, one obtains 27.9g of a white solid. Recrystallization from hot ethyl acetate and hexane afforded 22.3g (77% yield) of N-benzyloxycarbonyl-3 (S) -amino-1, 2 (S) -epoxy-4- phenylbutane, mp 102-103 °C artd MH+ 298.
Part C: A solution of N-benzyloxycarboriyl 3(S)-amino- 1,2-(S) -epoxy-4-phenylbutane (l.OOg, 3.36 mmol) and isoamylamine (4.90g, 67.2 mmol, 20 equiv.) in 10 mL of isopropyl alcohol was heated to reflux for 1.5 hours. The solution was cooled to room temperature, concentrated in vacuo and then poured into 100 mL of stirring hexane whereupon the product crystallized from solution. The product was isolated by filtration and air dried to give 1.18g, 95% of N=[[3(S)- phenylmethylcarbamoyl) amino-2 (R) -hydroxy-4- phenylbutyl]N-[ (3-methylbutyl) ] amine mp 108.0-109.5°C, MH+ m/z = 371.
EXAMPLE 20
Figure imgf000083_0001
Preparation of phenylmethyl f2R-hydroxy-3-f (3- methyIbutyl) (phenylsulfonyl) amino1 -IS-* (phenylmethyl) propyl]carbamate
From the reaction of N[3 (S) -benzyloxycarbonylamino- 2 (R) -hydroxy-4-phenyIbutyl] N-isoamylamine (1.47 gm, 3.8 mmol), triethylamine (528 uL, 3.8 mmol) and benzenesulfonyl chloride (483 uL, 3.8 mmol) one obtains phenylmethyl [2R-hydroxy-3-[ (3-meth Ibutyl) (phenylsulfonyl) amino] -IS- (phenylmethyl) propyl] - carbamate. Column chromotography on silica gel eluting with chloroform containing 1% ethanol afforded the pure product. Anal. Calcd for C29H36N205S: C, 66.39; H, 6.92; N, 5.34. Found: C, 66.37; H, 6.93; N, 5.26.
EXAMPLE 21
Figure imgf000084_0001
Preparation of 2R-hydroxy-3-[ [ (4-aminophenyl) sulfonyl] (2-methylpropyl) aminol -IS- (phenylmethylfpropylamine
Part A: Preparation of Carbamic acid, 2R-hydroxy-3- [ [ (4-nitrophenyl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl-, phenylmethyl ester
To a solution of 4.0g (10.8 mmol) of N-[3S-benzyloxy carbonylamino-2R-hydroxy-4-phenyl] -N-isobutylamine in 50mL of anhydrous methylene chloride, was added 4.5mL (3.27g, 32.4 mmol) of triethylamine. The solution was cooled to 0°C and 2.63g (11.9 mmol) of 4-nitrobenzene sulfonyl chloride was added, stirred for 30 minutes at 0°C, then for 1 hour at room temperature. Ethyl acetate was added, washed with 5% citric acid, saturated sodium bicarbonate, brine, dried and concentrated to yield 5.9g of crude material. This was recrystallized from ethyl acetate/hexane to afford 4.7g of pure carbamic acid, [2R-hydroxy-3-[ [ (4-nitrophenyl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester, m/e=556(M+H) .
Part B: Preparation of 2R-hydroxy-3- [[ (4-aminophenyl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propylamine A solution of 3.0g (5.4 mmol) of carbamic acid, 2R- hydroxy-3-[ [ (4-nitrophenyl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester in 20 mL of ethyl acetate was hydrogenated over 1.5g of 10% palladium-on-carbon catalyst under 35 psig of hydrogen for 3.5 hours. The catalyst was removed by filtration and the solution concentrated to afford 2.05g of the desired 2R-hydroxy-3- [ [ (4-aminophenyl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propylamine, m/e=392(M+H) .
EXAMPLE 22
Figure imgf000085_0001
Preparation of 2R-hvdroxy-3) [ (3- ammnophenyl) sulfonyl] (2-methylpropyl) aminol-lS- (phenylmethyl) propylamine
Part A: Preparation of Carbamic acid, [2R-hydroxy-3- [ (3-nitrophenylsulfonyl) ( 2-methylpropyl) amino] -1S- (phenylmethyl) propyl-, phenylmethyl ester
To a solution of l.lg (3.0 mmol) of N-[3S-benzyloxy carbonylamino-2R-hydroxy-4-phenyl]-N-isobutylamine in 15mL of anhydrous methylene chloride, was added 1.3mL (0.94g, 9.3 mmol) of triethylamine. The solution was cooled to 0°C and 0.67g (3.0 mmol) of 3-nitrobenzene sulfonyl chloride was added, stirred for 30 minutes at 0°C, then for 1 hour at room temperature. Ethyl acetate was added, washed with 5% citric acid, saturated sodium bicarbonate, brine, dried and concentrated to yield 1.74g of crude material. This was recrystallized from ethyl acetate/hexane to afford 1.40g of pure carbamic acid, [2R-hydroxy-3-[ (3-nitrophenylsulfonyl) (2- methylpropyl) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester, m/e=562 (M+Li) .
Part B: Preparation of [2R-hydroxy-3-[ [ (3- aminophenyl) sulfonyl] (2-methylpropyl) amino]-lS- (phenylmethyl) propylamine
A solution of 1.33g (2.5 mmol) of carbamic acid, [2R- hydroxy-3-[ (3-nitrophenylsulfonyl) (2- methylprop 1) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester in 40 mL of 1:1 methanol/tetrahydrofuran was hydrogenated over 0.70g of 10% palladium-on-carbon catalyst under 40 psig of hydrogen for 1.5 hours . The catalyst was removed by filtration and the solution concentrated to afford 0.87g of the desired [2R-hydroxy-3-[ [ (3-aminophenyl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propylamine.
EXAMPLE 23
Figure imgf000087_0001
Preparation of 2S-[ (pyrrolidin-l-yl) acetylamino]-N- [2R- hvdroxy-3-TN1- (2-methylpropyl) -N1- (2 , 3- dihydrobenzofuran-5-ylsulfonyl) amino] -1S- (phenylmethy1) propyl1 -3S-methylpentanamide
Part A: Preparation of pyrrolidineacetic acid hydrochloride
A solution of 19.6g (101 mmol) of t-butyl bromoacetate in 150 mL of THF was cooled in an ice bath and treated dropwise over about 0.5 hour with a solution of 14.4g
(202 mmol) of pyrrolidine in 75 mL of THF, to produce a white precipitate. The bath was removed and the reaction slurry stirred for two hours. The solid was removed by filtration and the filtrate was concentrated under reduced pressure to yield a clear liquid over an orange-colored solid. The liquid was cooled in an ice bath, then treated with 40 mL (80 mmol) of 4N HCl in dioxane and stirred for 15 hours. The solvents were removed in vacuo , and the residue was triturated with diethyl ether, then filtered to yield 12.9g of the desired acid as an off-white solid.
Part B: Preparation of 5- (2 , 3-dihydrobenzofuranyl) sulfonyl chloride To a solution of 3.35g of anhydrous N,N- dimethylformamide at 0°C under nitrogen was added 6.18g of sulfuryl chloride, whereupon a solid formed. After stirring for 15 minutes, 4.69g of 2 , 3-dihydrobenzofuran was added, and the mixture heated at 100°C for 2 hours. The reaction was cooled, poured into ice water, extracted with methylene chloride, dried over magnesium sulfate, filtered and concentrated the crude material. This was recrystallized from ethyl acetate to afford 2.45g of 5- (2 , 3-dihydrobenzofuranyl) sulfonyl chloride.
Part C: Preparation of Carbamic acid, 2R-hydroxy-3- [ [ (2, 3-dihydrobenzofuran-5-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester
To a solution of l.llg (3.0 mmol) of N-[3S-benzyloxy carbonylamino-2R-hydroxy-4-phenyl] -N-isobutylamine in 20mL of anhydrous methylene chloride, was added 1.3mL (0.94g, 9.3 mmol) of triethylamine. The solution was cooled to 0°C and 0.66g of 5- (2 , 3-dihydrobenzofuranyl) sulfonyl chloride was added, stirred for 15 minutes at 0°C, then for 2 hour at room temperature. Ethyl acetate was added, washed with 5% citric acid, saturated sodium bicarbonate, brine, dried and concentrated to yield 1.62g of crude material. This was recrystallized from diethyl ether to afford 1.17g of pure carbamic acid, [2R-hydroxy-3-[ [ (2 , 3-dihydrobenzofuran-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester.
Part D: Preparation of [2R-hydroxy-3-[ [ (2 , 3-dihydro benzofuran-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propylamine
A solution of 2.86g of carbamic acid, [2R-hydroxy-3-
[ [ (2 , 3-dihydrobenzofuran-5-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester in 30 mL of tetrahydrofuran was hydrogenated 0.99g of 10% palladium-on-carbon under 50 psig of hydrogen for 16 hours. The catalyst was removed by filtration and the filtrate concentrated to afford 1.99g of the desired
[2R-hydroxy-3-[ [ (2, 3-dihydrobenzofuran-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propylamine.
Part E: Preparation of 2S-[ (carbobenzyloxy) amino] -N- [2R-hydroxy-3-[ (3-methylpropyl) (2,3-dihydrobenzofuran-5- ylsulfonyl) amino] -IS- (phenylmethyl) prop l] -3S- methylpentanamide
A solution of 5.8g (22.0 mmol) of N-CBZ-L-isoleucine in 45 mL of anhydrous N,N-dimethylformamide (DMF) was cooled to 0°C and charged with 3.9g (28.7 mmol) of N- hydroxybenzotriazole (HOBT) and 4.2g (22.0 mmol) of 1- (3-dimethylaminopropy1) -3-ethylcarbodiimide hydrochloride (EDC) . The ice bath was removed after 20 minutes and stirring was continued for an additional 40 minutes. The reaction solution was then charged with a solution of 8.0g (19.1 mmol) of 2R-hydroxy-3-[ [ (2, 3- dihydrobenzofuran-5-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propylamine and 2.2g (22.0 mmol) of 4- methylmorpholine in 25 mL of anhydrous DMF and stirred for 15 hours. The solvents were removed in vacuo and the residue was partitioned between 300 mL of ethyl acetate and 120 mL of 5% potassium hydrogen sulfate solution. The layers were separated, and the organic layer was washed with 120 L each of saturated sodium bicarbonate solution, water and brine, then dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo to afford 16.7g of crude material. The crude material was crystallized from ethanol to yield 12. Og (94%) of 2S-[ (carbobenzyloxy) amino] -N-[2R-hydroxy-3-[ (3- methylpropyl) (2 , 3-dihydrobenzofuran-5- ylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S- methylpentanamide: m/e = 672 (M+Li) .
Part F: Preparation of 2S-amino-N-[2R-hydroxy-3-[ (3- methylpropyl) (2, 3-dihydrobenzofuran-5-ylsulfonyl) amino] - IS- (phenylmethyl) propyl] -3S-methylpentanamide
A Fischer-Porter bottle equipped with a magnetic stir bar was charged with 11.9g (17.9 mmol) of 2S- [ (carbobenzyloxy) amino] -N-[2R-hydroxy-3-[ (3- methylpropyl) (2 , 3-dihydrobenzofuran-5- * ylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S- meth lpentanamide and 75 mL of tetrahydrofuran (THF) . The solution was hydrogenated in the presence of 5g of 10% palladium-on-carbon catalyst (50% water by weight) under 50 psig of hydrogen for 4 hours at room temperature. The catalyst was removed by filtration, and the solvent removed in vacuo . The residue was dissolved in 300 mL of ethyl acetate and washed with 120 mL each of saturated sodium bicarbonate solution and brine, then dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to afford 8.8g of the desired product, m/e = 532 (M+H) .
Part G: Preparation of 2S-[ (pyrrolidin-l- yl) acetylamino] -N-[2R-hydroxy-3-[N1- (2-methylpropyl) -N1- (2 , 3-dihydrobenzofuran-5-ylsulfonyl) amino] -1S- (phenylmethyl) propyl] -3S-methylpentanamide
A solution of 3.7g (22.1 mmol) of pyrrolidineacetic acid hydrochloride in 45 L of anhydrous DMF was cooled to 0°C and charged with 3.4g (24.7 mmol) of HOBT and 3.6g (19.0 mmol) of EDC. The ice bath was removed after 20 minutes and stirring was continued for an additional 40 minutes. The reaction solution was then charged with a solution of 8.8g (16.5 mmol) of 2S-amino-N-[2R-hydroxy- 3- [ (3-methylpropyl) (2,3-dihydro benzofuran-5- ylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S- methylpentanamide and 4.5g (44.1 mmol) of 4- methylmorpholine in 25 mL of anhydrous DMF and stirred for 16 hours. The solvents were removed in vacuo and the residue was partitioned between 300 mL of ethyl acetate and 120 mL of 5% potassium hydrogen sulfate solution. The layers were separated, and the organic layer was washed with 120 mL each of saturated sodium bicarbonate solution, water and brine, then dried over anhydrous magnesium sulfate, filtered a"nd concentrated in vacuo to afford 9.9g of crude material. The coupling reaction was run again using 2.5g (15.1 mmol) of pyrrolidineacetic acid hydrochloride, 2.3g (17.0 mmol) of HOBT, 2.45g (12.8 mmol) of EDC, 3.0g (30.0 mmol) of 4-methylmorpholine, and the 9.9g of crude product in place of the amine from Part B. The reaction work-up was repeated and yielded 10.2g of crude product. Purification was accomplished using a Prep 2000 chromatograph on silica gel using 70-100% (5% methanol/95% ethyl acetate) /hexane to yield the desired product as a white solid, -m/e = 649 (M+Li) .
EXAMPLE 24
Figure imgf000092_0001
Preparation of N-[ (1, l-dimethylethoxyl)carbonyl]-N-[2- methylpropyl] -3S-[N1- (phenyImethoxycarbonyl) amino] -2R- hydroxy-4-phenylbutylamine
To a solution of 7.51g (20.3 mmol) of N-[3S- [ (phenylmethoxycarbony1) amino] -2R-hydroxy-4- phenylbutyl]-2-methylpropylamine in 67 mL of anhydrous tetrahydrofuran was added 2.25g (22.3 mmol) of triethylamine. After cooling to 0°C, 4.4g (20.3 mmol) of di-tert-butyldicarbonate was added and stirring continued at room temperature for 21 hours. The volatiles were removed in vacuo, ethyl acetate added, then washed with 5% citric acid, saturated sodium bicarbonate, brine, dried over magnesium sulfate, filtered and concentrated to afford 9.6g of crude product. Chromatography on silica gel using 30% ethyl acetate/hexane afforded 8.2g of pure N-[[3S- (phenylmethylcarbamoyl) amino] -2R-hydroxy-4-phenyl]-l- [ (2-methylpropyl) amino-2- (1,1- dimethylethoxyl) carbonyl]butane, mass spectrum m/e = 477 (M+Li) . EXAMPLE 25
Preparation of 2S-T [bromoacety11 amino1 -N-r2R-hvdroxy-3• fN1-(3-methyl-butyl) -N1- (phenylsulfonyl) aminol-lS- (phenylmethyl) propyll -3 , 3-dimethylbutaneamide
Figure imgf000093_0001
Part A: To a solution of N-CBZ-L-tert-leucine (450 mg, 1.7 mmol) and N-hydroxybenzotriazole (260 mg, 1.7 mmol) in DMF (10 mL) was added EDC (307 mg, 1.6 mmol). The solution was stirred for 60 minutes at room temperature and then 2R-hydroxy-3-[N-(3-methylbutyl) -N- (phenylsulfony1) amino] -IS- (phenylmethyl) propylamine (585 mg, 1.5 mmol) in DMF (2 mL) was added. The reaction was stirred for 16 hours at room temperature, then poured into a 50% saturated solution of sodium bicarbonate (200 mL) . The aqueous mixture was extracted thrice with ethyl acetate (50 mL) . The combined ethyl acetate layers were washed with water (50 mL) and saturated NaCl solution (50 mL) , then dried over magnesium sulfate. Filtration and concentration produced an oil which was chromatographed on silica gel (50 gm) eluting with 20% ethyl acetate in hexane. The phenylmethyl [lS-[ [ [2R-hydroxy-3-[ (3-methyIbutyl)
(phenylsulfonyl) amino] -IS- (phenylmethyl) propyl] amino] carbonyl] -2 , 2-dimethylpropyl] carbamate was obtained as a solid Anal. Calcd for C35H47N306S: C, 65.91; H, 7.43; N, 6.59. Found: C, 65.42; H, 7.24; N, 6.55. Part B: A solution of phenylmethyl [lS-[ [ [2R-hydroxy-3- [ (3-methyIbutyl) (phenylsulfonyl) -amino] -1S- (phenylmeth 1) propyl] amino] carbonyl] -2 , 2- dimethylpropyl]carbamate (200 mg, 0.31 mmol) in methanol (15 mL) was hydrogenated over 10% palladium on carbon for 2 hours. The reaction was filtered through diatomaceous earth and concentrated to an oil.
Part C: The resulting free amine from part B (150 mg, 0.3 mmol) was combined with diisopropylethylamine
(114 uL, 0.33 mmol) in dichloromethane \5 mL) . To this was added bromoacetyl chloride (27 uL, 0.33 mmol) dropwise. The reaction was stirred for 30 minutes at room temperature, then diluted with dichloromethane (30 mL) and extracted with 1 N HC1, water, and then saturated NaCl solution (25 mL each) . The organic solution was dried over MgS04 and concentrated to a solid. The 2S-[ [bromoacetyl] amino] -N-[2R-hydroxy-3-[ (3- methyIbutyl) (phenylsulfonyl) amino] -1S- (phenylmethyl) propyl] -3, 3-dimethylbutaneamide was sufficiently pure for use in the next step. This material can also be prepared by substituting bromoacetic anhydride for bromoacetyl chloride, or one can use chloroacetyl chloride or chloracetic anhydride.
EXAMPLE 26
Figure imgf000095_0001
Preparation of 2S-[chloroacetylaminol-N-r2R-hγdroxy-3- [N1- (2-methylbutyl)-N1- (phenylsulfonyl) aminol-lS- (phenylmethy1) ropyl1 -3S-methylpentanamide
Part A: To a solution of 2R-hydroxy-3-[N- (3- methyIbutyl) -N- (phenylsulfonyl) amino] -IS- (phenylmethyl) propylamine (2.79g, 7.1 mmol) in 27 mL of dioxane was added (2.3g, 7.1 mmol) of N-t-butylcarbonyl-L- isoleucine-N-hydroxysuccinamide ester, and the reaction was stirred under nitrogen atmosphere for 16 hours. The contents of the reaction were concentrated in vacuo , and the residue dissolved in ethyl acetate, washed with potassium hydrogen sulfate (5% aqueous) , saturated sodium bicarbonate, and saturated sodium chloride. The organic layer was dried over magnesium sulfate, filtered and concentrated to yield 4.3g of crude material which was chromatographed using 3:1 ethyl acetate: hexane to obtain 3.05g, 72% yield of Pentanamide, 2S-[[(1,1- dimethylethoxy) carbonyl] amino] -N- [ 2R-hydroxy-3- [ (3- methylbutyl) phenylsulfonyl) amino] -1S- (phenylmethyl) propyl] -3-methyl-.
Part B: (3.05g, 5.0 mmol) of the product from Part A was dissolved in 20 mL of 4N HC1 in dioxane and stirred under nitrogen atmosphere for 1.5 hours. The contents were concentrated in vacuo , and chased with diethyl ether. The crude hydrochloride salt was pumped on at 1 mm Hg until dry to yield 2.54g of product as its hydrochloride salt.
Part C: (2.54g, 5.0 mmol) of amine hydrochloride was dissolved in 50 mL of tetrahydrofuran and to this was added (l.Olg, 10 mmol) of 4-methyl-morpholine, at which time a precipitate forms. To this suspension was added chloroacetic anhydride (0.865g, 5.0 mmol) and stirred for 40 minutes. The contents were concentrated in vacuo, and the residue partitioned in ethyl acetate (200 mL) and 5% KHS04. The organic layer was washed with saturated sodium bicarbonate, and saturated sodium chloride, dried over magnesium sulfate, filtered and concentrated to yield the. crude product. Purification by silica gel chromatography using an eluant of 1:1 ethyl acetate: hexanes yielded 1.89g of pure chloroacetamide.
EXAMPLE 27
Figure imgf000097_0001
Preparation of 2S-T (pyrrolidin-l-yl) acetylamino1 -N- f2R-hvdroxy-3- \N1- (2-methylpropyl) -N1- (4-methoxyphenyl sulfonyl) amino] -1S- (phenylmethy1) propyl1 -3S-methylpentanamide
Part A: To a cooled solution of N-t-Boc-L-isoleucine 2.31g (10 mmol) and 2.00g (13.11 mmol) of N- hydroxybenzotriazole in 17 mL of N,N-dimethylformamide was added 1.91g (10 mmol) of EDC and stirred at 0°C for one hour. To this was added a solution of 4.0g (10 mmol) of 2R-hydroxy-3-[ (2-methylpropyl) (4- methoxyphenylsulfonyl) amino] -IS-(phenylmethyl) propylamine in 6 mL of N,N-dimethylformamide and the solution stirred for 16 hours. The solvent was removed by rotory evaporation, replaced with ethyl acetate, and washed with saturated sodium bicarbonate, 5% citric acid and brine. The organics were dried over magnesium sulfate, filtered and concentrated to yield 6.1g of crude product, which was chromatoraphed on silica gel using and eluant of 1:1 ethyl acetate:hexane to produce 5.12g (83% yield) of 2S-[ (tert-butoxycarbonyl) amino] -N- [2R-hydroxy-3-[ (3-methylpropyl) (4-methoxyphenylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S-methylpentanamide.
Part B: 5.00g (8.0 mmol) of the product from Part A was dissolved in 20 mL of 4N HC1 in dioxane and stirred for 20 minutes. The precipitated product was stripped two times from diethyl ether to yield 2S- (amino) -N-[2R- hydroxy-3-[ (3-methylpropyl) (4-methoxyphenylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S- methylpentanamide'hydrochloride, which was used in Part C without further purification.
Part C: The amine hydrochloride from part B was dissolved in 45 mL of methylene chloride and 3. Og N,N- diisopropylethylamme was added, followed by 1.22g (7.11 mmol) of chloroacetic anhydride. The solution was stirred at room temperature for 30 minutes. The contents were concentrated in vacuo and the residue was partitioned between ethyl acetate and water. The organic layer was washed with 5% citric acid, saturated sodium bicarbonate and brine. The organics were dried over magnesium sulfate filtered and concentrated in vacuo to yield 2S-[ (chloroacetyl) amino]-N-[2R-hydroxy-3- [ (3-methylpropyl) (4-methoxyphenylsulfonyl) amino] -1S- (phenylmethyl) propyl] -3S-methylpentanamide as a crude white foam, which was used in Part D without further purification.
Part D: 4.8g (8.0 mmol) of 2S-[ (chloroacetyl) amino] -N- [2R-hydroxy-3-[ (3-methylpropyl) (4-methoxyphenylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S-methylpentanamide was dissolved in 15 mL of tetrahydofuran and to this was added water followed by 2.8 mL (40 mmol) of pyrrolidine and the reaction stirred for 1.5 hours. The solvents were removed in vacuo and the residue was dissolved in ethyl acetate. The mixture was washed successively with saturated sodium bicarbonate and brine, dried over magnesium sulfate filtered and concentrated in vacuo to yield 5.6g of crude product. Purification by silica gel flash chromatograpy using an eluant of 1-3% methanol in dichloromethane yielded 3.2g of 2S-[ (pyrrolidin-1- yl) acetylamino]-N-[2R-hydroxy-3-[N1-(2-methylpropyl) -N1- (4-methoxyphenylsulfonyl) amino] -IS- (phenylmethyl) propyl] -3S-methylpentanamide as a white solid.
EXAMPLE 28
Figure imgf000099_0001
Preparation of Carbamic acid, 2R-hydroxy-3-[ [ (2- aminobenzothiazol-6-yl) suifonyll (2-methylpropyl) amino1 - IS- (phenylmethyl) propyl-, phenylmethyl ester
Carbamic acid, 2R-hydroxy-3-[ [ (4- aminophenyl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl-, phenylmethyl ester 0.30g (0.571 mmol) was added to a well mixed powder of anhydrous copper sulfate (1.20 g) and potassium thiocyanate (1.50 g) followed by dry methanol (6 mL) and the resulting black-brown suspension was heated at reflux for 2 hrs. The reaction mixture was filtered and the filtrate was diluted with water (5 mL) and heated at reflux. Ethanol was added to the reaction mixture, cooled and filtered. The filtrate upon concentration afforded a residue which was chromatographed (ethyl acetate:hexane 80:20) to afford 0.26g (78%) of the desired compound as a solid. EXAMPLE 29
Figure imgf000100_0001
Preparation of Carbamic acid, 2R-hydroxy-3-
[ fbenzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl-, phenylmethyl ester
Method 1: Carbamic acid, 2R-hydroxy-3-[ [ (2- aminobenzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl-, phenylmethyl ester (0.25g, 0.429 mmol) was added to a solution of isoamylnitrite (0.116 mL, 0.858 mmol) in dioxane (5 mL) and the mixture was heated at 85°C. After the cessation of evolution of nitrogen, the reaction mixture was concentrated and the residue was purified by chromatography (hexane: ethyl acetate 5:3) to afford 0.130g (53%) of the desired product as a solid.
Method 2: Crude benzothiazole-6-sulfonyl chloride in ethyl acetate (100 mL) was added to N-[3S- benzyloxycarbonyl amino-2R-hydroxy-4-phenyl] -N- isobutylamine (l.03g, 2.78 mmol) followed by N- methylmorpholine (4 mL) . After stirring at room temperature for 18 hr. , the reaction mixture was diluted with ethyl acetate (100 mL) , washed with citric acid (5%, 100 mL) , sodium bicarbonate (saturated, 100 mL) and brine (100 mL) , dried (MgS04) and concentrated in vacuo . The residue was chromatographed (silica gel, ethyl acetate: hexane 1:1) to afford 0.340g (23%) of desired product.
EXAMPLE 30
Figure imgf000101_0001
Preparation of Carbamic acid, 2R-hvdroxy-3- [ [ (2-amino benzothiazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl- , phenylmethyl ester; and Carbamic acid. 2R-hvdroxy-3-f r (2-aminobenzothiazol-7-yl) sulfonyll (2-methylpropyl) amino] -IS- (phenylmethyl) propyl- , phenylmethyl ester
The carbamic acid, 2R-hydroxy-3-[ (3-aminophenylsulfonyl) (2-methylpropyl) amino] -IS- (phenylmethyl) propyl-, phenylmethyl ester 0.36g (0.685 mmol) was added to a well mixed powder of anhydrous copper sulfate (1.44 g) and potassium thiocyanate (1.80 g) followed by dry methanol (10 mL) and the resulting black-brown suspension was heated at reflux for 2 hrs. The reaction mixture was filtered and the filtrate was diluted with water (5 mL) and heated at reflux. Ethanol was added to the reaction mixture, cooled and filtered. The filtrate upon concentration afforded a residue which was chromatographed (ethyl acetate:hexane 1:1) to afford O.lδg (45%) of the 7-isomer as a solid. Further elution of the column with (ethyl acetate:hexane 3:2) afforded 0.80g (20%) afforded the 5-isomer as a solid.
EXAMPLE 31
Figure imgf000102_0001
Preparation of 3S-amino-l-rN-(2-methylpropyl) -N-(4- methoxyphenylsulfonyl) amino]-4-phenyl-2R-butanol
Part A: N-benzyloxycarbonyl-3 (S) -amino-l-chloro-4- pheny1-2 (S) -butanol
To a solution of N-benzyloxycarbonyl-L-phenylalanine chloromethyl ketone (75g, 0.2 mol) in a mixture of
800 mL of methanol and 800 mL of tetrahydrofuran was added sodium borohydride (13.17g, 0.348 mol, 1.54 equiv.) over 100 min. The solution was stirred at room temperature for 2 hours and then concentrated in vacuo . The residue was dissolved in 1000 mL of ethyl acetate and washed with IN KHS04, saturated aqueous NaHC03 , saturated aqueous NaCl, dried over anhydrous MgS04, filtered and concentrated in vacuo to give an oil. The crude product was dissolved in 1000 mL of hexanes at 60°C and allowed to cool to room temperature where upon crystals formed that were isolated by filtration and washed with copious amounts of hexanes. This solid was then recrystallized from hot ethyl acetate and hexanes to provide 32.3g 43% of N-benzyloxycarbonyl-3 (S) -amino- l-chloro-4-phenyl-2(S)-butanol, mp 150-151°C, FAB MS: MLi+ = 340.
Part B: 3 (S) -[N- (benzyloxycarbonyl) amino]-l, 2 (S) -epoxy- 4-phenylbutane
A solution of potassium hydroxide (6.52g, 0.116 mol, 1.2 equiv.) in 970 mL of absolute ethanol was treated with N-benzyloxycarbonyl-3 (S) -amino-l-chloro-4-phenyl-2 (S) - butanol (32.3g, 0.097 mol). This solution was stirred at room temperature for 15 minutes and then concentrated in vacuo to give a white solid. The solid was dissolved in dichloromethane and washed with water, dried over anhyd MgS04, filtered and concentrated in vacuo to give a white solid. The solid was crystallized from hexanes and ethyl acetate to give 22.3g, 77% of 3(S)-[N-
(benzyloxycarbonyl) amino]-l, 2 (S) -epoxy-4-phenylbutane, mp 102-103°C, FAB MS: MH+ = 298.
Part C: N- [ 3 (S) -benzyloxycarbonylamino-2 (R) -hydroxy-4- phenyl]N-isobutylamine
A solution of N-benzylcarbonyl-3 (S) -amino-1, 2- (S) -epoxy- 4-phenyl butane (50. Og, 0.168 mol) and isobutylamine (246g, 3.24 mol, 20 equivalents) in 650 mL of isopropyl alcohol was heated to reflux for 1.25 hours. The solution was cooled to room temperature, concentrated in vacuo and then poured into 1 L of stirring hexane whereupon the product crystallized from solution. The product was isolated by filtration and air dried to give 57.56g, 92% of N [ 3 (S) -benzyloxycarbonylamino-2 (R) - hydroxy-4-phenyl]-N-isobutylamine, mp 108.0-109.5°C, MH+ m/z=371.
Part D: phenylmethyl [2 (R) -hydroxy-3-[N- (2- methylpropyl) -N- (4-methoxyphenylsulfonyl) amino] -1S- (phenylmethyl) propyl] carbamate
The amine from Part C (936.5 mg, 2.53 mmol) and triethylamine (2.88.5 mg, 2.85 mmol) was dissolved in 20 mL of dichloromethane and treated with 4- methoxybenzenesulfonyl chloride (461 mg, 2.61 mmol). The solution was stirred at room temperature for 16 hours and then concentrated in vacuo . The residue was dissolved in ethyl acetate and this solution was washed with IN KHS0 , saturated aqueous NaHC03, brine, dried over anhyd MgS04, filtered, and concentrated to give a clear oil 1.234g. The oil was crystallized from a mixture of ether and hexanes, 729.3 mg, 56.5% mp 95- 99°C, FAB MS: MH+ = 511.
Part E: 3S-amino-l-[N-(2-methylpropyl) -N-(4- methoxyphenylsulfonyl) amino] -4-phenyl-2R-butanol A solution of phenylmethyl [2 (R) -hydroxy-3-[N-(2- methylpropyl) -N- (4-methoxyphenylsulfonyl) amino] 1-S- (phenylmethyl) propyl carbamate (671.1 mg, 1.31 mmol) from Part D in 10 mL of methanol was hydrogenated over 50 mg of 10% palladium on carbon at 40 psig at room temperature for 15 hours. The catalyst was removed by filtration through diatomaceous earth and the filtrate concentrated to give a white foam, 474.5 mg, 96%, FAB MS: MH+ = 377. EXAMPLE 32
Figure imgf000105_0001
Preparation of 1 , 3-benzodioxole-5-sulfonyl chloride
Method 1: To a solution of 4.25g of anhydrous N,N- dimethylformamide at 0°C under nitrogen was added 7.84g of sulfuryl chloride, whereupon a solid formed. After stirring for 15 minutes, 6.45g of 1, 3-benzodioxole was added, and the mixture heated at 100°C for 2 hours. The reaction was cooled, poured into ice water, extracted with methylene chloride, dried over magnesium sulfate, filtered and concentrated to give 7.32g of crude material as a black oil. This was chromatographed on silica gel using 20% methylene chloride/hexane to afford 1.9g of (l,3-benzodioxol-5-yl) sulfonyl chloride.
Method 2: To a 22 liter round bottom flask fitted with a mechanical stirrer, a cooling condenser, a heating mantle and a pressure equalizing dropping funnel was added sulfur trioxide DMF complex (2778g, 18.1 moles). Dichloroethane (4 liters) was then added and stirring initiated. 1,3-Benzodioxole (1905g, 15.6 moles) as then added through the dropping funnel over a five minute period. The temperature was then raised to 75°C and held for 22 hours (NMR indicated that the reaction was done after 9 hours.) The reaction was cooled to 26° and oxalyl chloride (2290g, 18.1 moles) was added at a rate so as to maintain the temperature below 40°C (1.5 hours) . The mixture was heated to 67 °C for 5 hours followed by cooling to 16 °C with an ice bath. The reaction was quenched with water (51) at a rate which kept the temperature below 20°C. After the addition of water was complete, the mixture was stirred for 10 minutes. The layers were separated and the organic layer was washed again twice with water (51) . The organic layer was dried with magnesium sulfate (500g) and filtered to remove the drying agent. The solvent was removed under vacuum at 50°C. The resulting warm liquid was allowed to cool at which time a solid began to form. After one hour, the solid was washed with hexane (400 L) , filtered and dried to provide the desired sulfonyl chloride (2823g) . The hexane wash was concentrated and the resulting solid washed with 400 mL hexane to provide additional sulfonyl chloride (464g) . The total yield was 3287g (95.5% based upon 1,3- benzodioxole) .
Method 3: Dichloroethane (DCE) , 2 liters, was added to a 30 L reactor fitted with condenser (0°C) , two agitator mechanical stirrer, internal cooling coils heating mantel and nitrogen atmosphere/purge system. Sulfur trioxide«DMF complex (4856g, 31.7 moles) was then added over a 15 minute period. No temperature rise was observed. The reaction mixture was heated to 75°C. The material was completely dissolved in two hours. NMR analysis indicated that the reaction was complete in less than 5 hours. The reaction was allowed to cool overnight for convenience. The reaction mass was cooled to 15°C. A caustic scrubber (a 4L Erlenmeyer flask containing 2 L of 25% NaOH with subsurface addition) was attached to the top of the condenser. (The scrubber was changed when NaCl precipitation was significant) .
Oxalyl chloride (4041g, 31.84 moles) was added over a 1.5 hour period while maintaining the temperature between 11° and 15°C. The reaction mixture was allowed to come to room temperature over 30 minutes at which time gas evolution had ceased. NMR analysis after an additional hour at room temperature indicated that the reaction was complete. The reaction mass was then cooled to 15°C and water (2 L) was added very slowly while keeping the temperature below 25°C. CAUTION: the initial addition of water is very exothermic. The first 50 mL of water caused the temperature to rise to 25°C. After the initial 2 L of water was added, an additional 6 L was then added quickly. The reaction mixture was stirred for 10 minutes. The stirring was stopped and the layers were allowed to separate. The water layer was removed and the layers allowed to separate. The water layer was removed and the organic" layer was washed twice with water (8 L) . The organic layer was then dried over magnesium sulphate (1 Kg) which clarified the solution. The solution was filtered through a sintered glass funnel in two batches and the residue from each was washed with DCE (1 L) . The solvent was removed at 50°C in vacuo (ca. 100 mm of Hg to remove the majority of the solvent then 2 mm of Hg) . The material solidified as it cooled to room temperature to yield the desired sulfonyl chloride (5337g, 91.65% yield) which was used in the subsequent step without further purification.
Method 4: l,4-benzodioxan-6-sulfonyl chloride was prepared according to the procedure disclosed in EP 583960, incorporated herein by reference.
EXAMPLE 33
Figure imgf000108_0001
Preparation of 1-fN-r (l,3-benzodioxol-5*-yl) sulfonyl]-N-(2-methylpropyl) amino1-3 (S) -
[bis (phenylmethyl) amino] -4-phenyl-2 (R) -butanol
Method 1: To a 5000 mL, 3-necked flask fitted with a mechanical stirrer was added N-[3 (S) -[N,N- bis (phenylmethyl) amino] -2 (R) -hydroxy-4-phenylbuty1] -N- isobutylamine»oxalic acid salt (354.7g, 0.7 mole) and 1,4-dioxane (2000 mL) . A solution of potassium carbonate (241.9g, 1.75 moles) in water (250 mL) was then added. The resultant heterogeneous mixture was stirred for 2 hours at room temperature followed by the addition of 1, 3-benzodioxole-5-sulfonyl chloride (162.2g, 0.735 mole) dissolved in 1,4-dioxane (250 mL) over 15 minutes. The reaction mixture was stirred at room temperature for 18 hours. Ethyl acetate (1000 mL) and water (500 mL) were charged to the reactor and stirring continued for another 1 hour. The aqueous layer was separated and further extracted with ethyl acetate (200 mL) . The combined ethyl acetate layers were washed with 25% brine solution (500 L) and dried over anhydrous magnesium sulfate. After filtering and washing the magnesium sulfate with ethyl acetate (200 mL) , the solvent in the filtrate was removed under reduced pressure yielding the desired sulfonamide as an viscous yellow foamy oil (440.2g 105% yield) HPLC/MS (electrospray) (m/z 601 [M+H]+].
EXAMPLE 34
Figure imgf000109_0001
Preparation of 1-fN-r (1, 3-benzodioxol-5-yl) sulfonyl]-N-(2-methylpropyl) amino1-3 (S) -amino-4-pheny1-
2 (R) -butanol*methanesulfonic acid salt
Method 1: Crude l-[N-[ (1, 3-benzodioxol-5-yl) sulfonyl] - N- (2-methylpropyl) amino] -3 (S) - [bis (phenylmethyl) amino] - 4-phenyl-2 (R) -butanol (6.2g, 0.010 moles) was dissolved in methanol (40 mL) . Methanesulfonic acid (0.969g, 0.010 moles) and water (5 mL) were then added to the solution. The mixture was placed in a 500 mL Parr hydrogenation bottle containing 20% Pd(OH)2 on carbon (255 mg, 50% water content) . The bottle was placed in the hydrogenator and purged 5 times with nitrogen and 5 times with hydrogen. The reaction was allowed to proceed at 35 °C with 63 PSI hydrogen pressure for 18 hours. Additional catalyst (125 mg) was added and, after purging, the hydrogenation continued for and additional 20 hours. The mixture was filtered through celite which was washed with methanol (2 X 10 mL) . Approximately one third of the methanol was removed under reduced pressure. The remaining methanol was removed by aziotropic distillation with toluene at 80 torr. Toluene was added in 15, 10, 10 and 10 mL portions. The product crystallized from the mixture and was filtered and washed twice with 10 mL portions of toluene. The solid was dried at room temperature at 1 torr for 6 hours to yield the amine salt (4.5g, 84%). HPLC/MS (electrospray) was consistent with the desired product (m/z 421 [M+H] +) .
Method 2:
Part A: N-[3 (S) -[N,N-bis (phenylmethyl) amino] -2 (R)- hydroxy-4-phenylbutyl]-N-isobutylamine»όxalic acid salt (2800g, 5.53 moles) and THF (4L) were added to a 22L round bottom flask fitted with a mechanical stirrer. Potassium carbonate (1921g, 13.9 moles) was dissolved in water (2.8L) and added to the THF slurry. The mixture was then stirred for one hour. l,3-benzodioxole-5- sulfonyl chloride (1281g, 5.8 moles) was dissolved in THF (1.4L) and added to the reaction mixture over 25 minutes. An additional 200 mL of THF was used to rinse the addition funnel. The reaction was allowed to stir for 14 hours and then water (4 L) was added. This mixture was stirred for 30 minutes and the layers allowed to separate. The layers was removed and the aqueous layer washed twice with THF (500 mL) . The combined THF layers were dried with magnesium sulfate (500 g) for one hour. This solution was then filtered to remove the drying agent and used in subsequent reactions.
Part B: To the THF solution of crude 1-[N-[(1,3- benzodioxol-5-y1) sulfonyl] -N- (2-methylpropyl) amino] - 3 (S) -[bis (phenylmethyl) amino] -4-phenyl-2 (R) -butanol was added water (500 mL) followed by methane sulfonic acid (531g, 5.5 moles). The solution was stirred to insure complete mixing and added to a 5 gallon autoclave. Pearl an's catalyst (200g of 20% Pd(0H)2 on C/ 50% water) was added to the autoclave with the aid of THF (500 mL) . The reactor was purged four times with nitrogen and four times with hydrogen. The reactor was charged with 60 psig of hydrogen and stirring at 450 rpm started. After 16 hours, HPLC analysis indicated that a small amount of the mono-benzyl intermediate was still present. Additional catalyst (50g) was added and the reaction was allowed to run overnight. The solution was then filtered through celite (500g) to remove the catalyst and concentrated under vacuum in five portions. To each portion, toluene (500 mL) was added and removed under vacuum to azeotropically removed residual water. The resulting solid was divided into three portions and each washed with methyl t-butyl ether (2 L) and filtered. The residual solvent was removed at room temperature in a vacuum oven at less than 1 torr to yield the 2714g of the expected salt.
If desired, the product can be further purified by the following procedure. A total of 500 mL of methanol and 17Og of material from above was heated to reflux until it all dissolved. The solution was cooled, 200 mL of isopropanol added and then 1000-1300 mL of hexane, whereupon a white solid precipitated. After cooling to 0°C, this precipitate was collected and washed with hexane to afford 123g of the desired material. Through this procedure, the original material which was a 95:5 mixture of alcohol diastereomers was greater than 99:1 of the desired diastereomer. EXAMPLE 35
Figure imgf000112_0001
Preparation of 2R-hydroxy-3-f f (1, 3-benzodioxol-5- yl) sulfonyl] (2-methylpropyl) amino] -1S- ( henylmethyl) propylamine
Part A: Preparation of 2R-hydroxy-3- [ [ (1, 3-benzodioxol- 5-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propylcarbamic acid phenylmethyl ester
To a solution of 3.19 g(8.6 mmol) of N-[3S-benzyloxy carbonylamino-2R-hydroxy-4-phenyl] -N-isobutylamine in 40mL of anhydrous methylene chloride, was added 0.87g of triethylamine. The solution was cooled to 0°C and 1.90g of (l,3-benzodioxol-5-yl) sulfonyl chloride was added, stirred for 15 minutes at 0°C, then for 17 hours at room temperature. Ethyl acetate was added, washed with 5% citric acid, saturated sodium bicarbonate, brine, dried and concentrated to yield crude material. This was recrystallized from diethyl ether/hexane to afford 4.77g of pure 2R-hydroxy-3-[ [ (1, 3-benzodioxol-5- yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propylcarbamic acid phenylmethyl ester.
Part B: Preparation of 2R-hydroxy-3-[ [ (1, 3-benzodioxol- 5-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propylamine A solution of 4.11g of carbamic acid, 2R-hydroxy-3- [ [ (l,3-benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl-, phenylmethyl ester in 45 mL of tetrahydrofuran and 25 mL of methanol was hydrogenated over l.lg of 10% palladium-on-carbon under 50 psig of hydrogen for 16 hours. The catalyst was removed by filtration and the filtrate concentrated to afford 1.82g of the desired 2R-hydroxy-3-[ [ (1, 3-benzodioxol-5- yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propylamine.
EXAMPLE 36
Figure imgf000113_0001
Preparation of Benzothiazole-6-sulfonyl Chloride
Part A: Preparation of N- (4-Sulfonamidophenyl) thiourea
A mixture of sulfanilamide (86g, 0.5 mole), ammonium thiocyanate (76. Og, 0.5 mole) and dilute hydrochloric acid (1.5 N, 1 L) was mechanically stirred and heated at reflux for 2 hr. About 200 mL of water was distilled off and concentration of the reaction mixture afforded a solid. The solid was filtered and was washed with cold water and air dried to afford 67.5g (59%) of the desired product as a white powder.
Part B: Preparation of 2-Amino-6- sulfonamidobenzothiazole
Bromine (43.20g, 0.27 mol) in chloroform (200 mL) was added over 1 hr. to a suspension of N-(4- sulfonamidophenyl) -thiourea (27.72, 0.120 mol) in chloroform (800 mL) . After the addition, the reaction mixture was heated at reflux for 4.5 hr. The chloroform was removed in vacuo and the residue was repeatedly distilled with additional amounts of chloroform. The solid obtained was treated with water (600 mL) followed by ammonium hydroxide (to make it basic) , then was heated at reflux for 1 hr. The cooled reaction mixture was filtered, washed with water and air dried to afford 22.0g (80%) of the desired product as a white powder.
Part C: Preparation of Benzothiazole-ϊ-sulfonic acid
A suspension of 2-amino-6-sulfonamido-benzothiazole (10. Og, 43.67 mmol) in dioxane (300 mL) was heated at reflux. Isoamylnitrite (24 mL) was added in two portions to the reaction mixture. Vigorous evolution of gas was observed (the reaction was conducted behind a shield as a precaution) and after 2 hr. , a red precipitate was deposited in the reaction vessel. The reaction mixture was filtered hot, and the solid was washed with dioxane and was dried. The solid was recrystallized from methanol-water. A small amount of a precipitate was formed after 2 days. The precipitate was filtered off and the mother liquor was concentrated in vacuo to afford a pale red-orange solid (8.0g, 85%) of pure product.
Part D: Preparation of 6-Chlorosulfonylbenzothiazole
Thionyl chloride (4 mL) was added to a suspension of the benzothiazole-6-sulfonic acid (0.60g, 2.79 mmol) in dichloroethane (15 mL) and the reaction mixture was heated at reflux and dimethylformamide (5 mL) was added to the reaction mixture to yield a clear solution. After 1.5 hr. at reflux, the solvent was removed in vacuo and excess HCl and thionyl chloride was chased by evaporation with dichloroethane.
EXAMPLE 37
Figure imgf000115_0001
Preparation of N-f2R-hydroxy-3-f f (1 , 3-benzodioxol-5- yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyll-2S-f f (pyrrolidin-l-yl) acetyl] aminol -3 ,3- dimethylbutanamide
Part A: Preparation of N-f2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S- [ (phenyImethoxycarbony1) amino] 3, 3-dimethylbutanamide
Figure imgf000115_0002
To a solution of 118.8g (0.776 mol) of N- hydroxybenzotriazole and 137. lg (0.52 mol) of N- carbobenzyloxycarbonyl-L-tert-leucine in 750 mL of anhydrous DMF at 0°C under a nitrogen atmosphere, was added 109. lg (0.57 mol) of EDC. After stirring at 0°C for 2 hours, a solution of 273g (0.53 mol) of 2R- hydroxy-3-[ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propylamine methanesulfonate, previously neutralized with 228 mL (210g, 2.08 mol) of 4-methylmorpholine, in 250 mL of anhydrous DMF was added. After stirring at 0°C for 30 minutes, the mixture stirred at room temperature for 18 hours. The solvents were removed under reduced pressure at 45°C, 1.5 L of ethyl acetate added, washed with 5% citric acid, saturated sodium bicabonate, brine, dried over anhydrous magnesium sulfate, filtered and concentrated to afford 400g of crude material. This was chromatographed in 3 batches on a Prep 2000 Chromatogram on silica gel using 20%-50% ethyl acetate/hexane as eluent to yield 320g of purified material, m/e=674 (M+Li) , 98% by HPLC.
Part B: Preparation of N-[2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-y1) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-amino-3 , 3-dimethylbutanamide
Figure imgf000116_0001
A solution of 312g of the Cbz compound from above in IL of tetrahydrofuran was hydrogenated in the presence of lOOg of 4% palladium-on-carbon catalyst under 60 psig of hydrogen for 6 hours at room temperature. The catalyst was removed by filtration and the solvents removed under reduced pressure to afford 240g of the desired compound.
Part C: Preparation of N- [2R-hydroxy-3- [ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl] -2S-[ (chloroacetyl) amino] -3 , 3- dimethylbutanamide
Figure imgf000117_0001
To a solution of 234.3g (0.439 mol) of the amine from above in IL of methylene chloride, was added 80 L (59.5g, 0.46 mol) of diisopropylethylamine, followed by the slow addition at room temperature of 78.8g (0.46 mol) of chloroacetic anhydride while maintaining the temperature below 35°C. After stirring for an additional 1 hour, analysis by HPLC indicated a small amount of starting material was still present, and 1.5g of chloroacetic anhydride was added. After 10 minutes, the solvents were removed under reduced pressure, 1 L ethyl acetate added, washed with 5% citric acid, saturated sodium bicarbonate, brine, dried over anhydrous magnesium sulfate, filtered and concentrated to yield 314g of crude material. This was chromatographed in 3 portions on a Prep 2000 Chromatogram on silica gel using 20-50% ethyl acetate/hexane to afford 165g of the desired compound, m/e=616 (M+Li) , 98% by HPLC.
Part D: Preparation of N- [2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl] -2S-[ [ (pyrrolidin-l- yl) acetyl]amino] -3, 3-dimethylbutanamide
Figure imgf000118_0001
To 164.2g (0.27 mol) of chloroacetyl compound from above was added 500 mL of tetrahydrofuran, the solvent removed under reduced pressure to remove any ethyl acetate, and then 350 mL of tetrahydrofuran was added. To this solution at 10°C was added 130 mL (1.56 mol) of pyrrolidine. After 1 hour, the solvents were removed under reduced pressure, 1 L ethyl acetate added, washed with saturated sodium bicarbonate, brine, dried over anhydrous magnesium sulfate, filtered and concentrated to afford 185g of crude material, which was assayed by HPLC to be 98.9% purity. This was split into 3 portions and chromatographed on a Prep 2000 Chromatogram using first 50% ethyl acetate/hexane, followed by 5% methanol/ethyl acetate to afford 160g of purified material (99% by HPLC) . This was then recrystallized from 460 mL of diethyl ether and 70 mL of hexane to afford 12lg of the desired product (>99% by HPLC) , m/e=651(M+Li) , mp=112-114°C.
EXAMPLE 38
Preparation of N-r2R-hydroxy-3-f f (1, 3-benzodioxol-5- yl) sulfonyll (2-methylpropyl) amino] -1S- (phenylmethyl) propyl1 -2S-f f (pyrrolidino) acetyl]amino]' 3S-methylpentanamide
Figure imgf000119_0001
Part A: Preparation of N-[2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl] -2S-[ [ (1, 1-dimethylethoxy) carbonyl] amino] -3S-methylpentanamide
Figure imgf000119_0002
To a cooled solution of N-t-Boc-L-isoleucine 2.02g (8.74 mmol) and 2.00g (13.11 mmol) of N-hydroxybenzotriazole in 17 mL of N,N-dimeth Iformamide was added 1.84g (9.61 mmol) of EDC and stirred at 0°C for one hour. To this was added a solution of 3.67g ( 8.74 mmol) of 2R- hydroxy-3- [ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl amine in 6 mL of N,N-dimethylformamide and the solution stirred for 16 hours. The solvent was removed in vacuo , replaced with ethyl acetate, and washed with saturated sodium bicarbonate, 5% citric acid and brine. The organic layers were dried over magnesium sulfate, filtered and concentrated to yield 6. lg of crude product, which was chromatoraphed on silica gel using 1:1 ethyl acetate:hexane eluant to produce 4.3g (78% yield) of N- [2R-hydroxy-3-[ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ [ (1,1- dimethylethoxy) carbonyl] amino] -3S-methylpentanamide.
Part B: Preparation of N-[2R-hydroxy-3-f [ (1/3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl ] -2S-amino-3S- methylpentanamide»hydrochloride salt
Figure imgf000120_0001
N-[2R-hydroxy-3-[ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-[ [ (1,1- dimethylethoxy) carbonyl] amino] -3S-methylpentanamide (4.29g, 6.77 mmol) was dissolved in 20 mL of 4N HCl in dioxane and stirred for 20 minutes. The precipitated product was stripped two times from diethyl ether and the crude hydrochloride salt was used in subsequent reactions .
Part C: Preparation of N-[2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl] -2S-[ (chloroacetyl) amino] -3S- methylpentanamide
Figure imgf000121_0001
N-[2R-hydroxy-3-[ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-amino- 3S-methylpentanamide»hydrochloride salt^(3.62g, 6.77 mmol) was dissolved in 45 mL of methylene chloride and to this was added 1.3g (10.15 mmol) of N,N- diisopropylethyl amine to neutralize the salt, and another 0.923g (7.10 mmol) of diisopropylethyl amine followed by 1.22g (7.11 mmol) of chloroacetic anhydride. The solution was stirred at room temperature for 30 minutes. The contents were concentrated on a rotory evaporator and the residue was partitioned between ethyl acetate and water. The organic layer was washed with 5% citric acid and then saturated sodium bicarbonate and brine. The organic layers were dried over magnesium sulfate filtered and concentrated to yield 4.12g of crude product. Recrystallization from ethyl acetate hexane yielded 3.5g (85% yield) of N-[2R-hydroxy-3- [[ (1, 3-benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -2S-[ (chloroacetyl) amino] -3S- methylpentanamide , as a white solid; mass spectrum m/z=616 (M+Li) .
Part D: Preparation of N- [2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl-amino] -1S- (phenylmethy1) propyl] -2S- [ [ (pyrrolidino) acetyl] amino] - 3S-methylpentanamide
Figure imgf000122_0001
N-[2R-hydroxy-3-[ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ (chloroacetyl) amino] -3S-methylpentanamide (5.18g, 8.49 mmol) was dissolved in 15 mL of tetrahydrofuran and to this was added 0.5 mL of water followed by 3.62g (50.9 mmol) of pyrrolidine and the reaction stirred for 1.5 hours. The solvents were removed by rotory evaporation and replaced with ethyl acetate. The solution was washed successively with saturated sodium bicarbonate and brine, dried over magnesium sulfate, filtered and concentrated to yield 5.6g of crude product. Purification by silica gel flash chromatograpy using an eluant of 1-3% methanol in dichloromethane yielded 3.8g of N-[2R-hydroxy-3-
[ [ (l,3-benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-l- yl) acetyl]amino] -3S-methylpentanamide as a white solid.
EXAMPLE 39
Preparation of N-r f2R-hvdroxy-3-f (1.3-benzodioxol-5- yl) sulfonyll (2-methylpropyl) aminol -IS- (plenylmetlyl) propyll-2S-f f (pyrrolidin-l-yl) acetyllaminol -3- methylbutaneamide
Figure imgf000123_0001
Part A : Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-y1) sulfonyl] (2-methylpropyl) amino]-1S- (phenylmethyl) propyl] -2S- [ (phenyImethoxycarbonyl) amino] 3-methylbutaneamide
Figure imgf000123_0002
A 250 mL round bottom flask equipped with magnetic stir bar was charged with N-Cbz-L-Valine (4.22g, 16.8 mmol) in 20 mL DMF. The solution was cooled to 0 C and charged with HoBt (2.96, 21.9 mmol) and EDC (3.22g, 16.8 mmol) and stirred 1 hour. The reaction was then charged with N-methylmorpholine (1.7g, 16.8 mmol), 2R-hydroxy-3- [ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propylamine (7.55g, 14.6 mmol) in 30 mL of DMF. The reaction was stirred overnight at room temperature then concentrated in vacuo and partioned between ethyl acetate and 5% Citric acid. The combined organic layers were washed with saturated sodium bicarbonate and brine, and dried over sodium sulfate. Concentration in vacuo yielded lOg crude product. Purification by Prep HPLC (20-40% ethyl acetate/hexane) yielded 5.8g (61 %) of the desired compound. Part B : Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino]-lS- (phenylmethy1) propyl] -2S-amino-3-methylbutaneamide
Figure imgf000124_0001
A 300 mL Fisher-Porter vessel equipped with magnetic stir bar was charged with N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S-
(phenylmethy1) propyl] -2S-[ (phenylmethoxycarbonyl) amino] - 3-methylbutaneamide (5.8 g) , 2.3g of 10% Pd-C in 75 mL tetrahydrofuran. The reaction was charged with 50 psi H2 and hydrogenated overnight. The reaction mixture was filtered thru Celite and concentrated in vacuo to yield 4.4g of white foam that was used in subsequent reactions without further purification.
Part C : Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S- [ (chloroacetyl) amino] -3- methylbutaneamide
Figure imgf000124_0002
A 250 L round bottom flask equipped with magnetic stir bar was charged with crude N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-amino-3-methylbutaneamide (4.35 g) in 60 mL CH2C12. The reaction was charged with 1.19g diisopropylamine followed by 1.5g of chloroacetic anhydride and stirred until TLC indicated no remaining starting material (about 1.5 hours). The reaction was concentrated in vacuo and partioned between ethyl acetate and saturated sodium bicarbonate. The combined organic layers were washed with brine, and dried over sodium sulfate. Concentration in vacuo yielded 5.17g of desired product that was used in subsequent reactions without further purification.
Part D : Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-l- yl) acetyl] amino] -3-methylbutaneamide
Figure imgf000125_0001
A 250 mL round bottom flask equipped with magnetic stir bar was charged with crude N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (plenylmetlyl) propyl ]-2S-[ , chloroacetyl) amino] -3- methylbutaneamide (4.99 g) and 3.57g pyrrolidine in 20 mL tetrahydrofuran and 0.5 mL H 0. After 45 minutes at room temperature, TLC analysis indicated complete reaction. The reaction was concentrated in vacuo and partioned between ethyl acetate and saturated sodium bicarbonate. The combined organic layers were washed with brine and dried over sodium sulfate. Concentration in vacuo yielded 5.2g crude product. Purification by Prep HPLC (3 % MeOH/ 57 % ethyl acetate/40 % hexane) gave 4.3g pure N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5- yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl ]-2S-[ [ (pyrrolidin-l-yl) acetyl ] amino] -3- methylbutaneamide .
EXAMPLE 40
Figure imgf000126_0001
Preparation of N-r r2R-hydroxy-3-f (4-methoxyphenyl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl1 -2S-f f (pyrrolidin-l-yl) acetyl] amino1-3- methylbutaneamide
To a solution of 2.4g N-[ [2R-hydroxy-3-[ (4- methoxyphenyl) sulfonyl] (2-methylpropyl) amino]-lS- (phenylmethyl) propyl] -2S- [ (chloroacetyl) amino] -3- methylbutaneamide in 65 mL THF was added 0.68 mL (2.0 eq.) of pyrrolidine and the reaction mixture was stirred at room temperature overnight. The reaction was concentrated in vacuo and partitioned between ethyl acetate and saturated aqueous sodium bicarbonate. The combined organics were washed with brine, dried, and concentrated in vacuo to a white foam. Trituration with diethyl ether yielded l.Og product (98 % pure by HPLC). EXAMPLE 41
Figure imgf000127_0001
Preparation of N-[2R-hydroxy-3-[ (phenylsulfonyl) (2- methylpropyl) amino] -IS- (phenylmethyl) prppyl] -2S-
[ [ (pyrrolidin-l-yl) acetyl] amino] -3S-methylpentanamide
Part A: Preparation of N-[2R-hydrσxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-[ [ (phenylmethoxy) carbonyl] amino] -3S-methylpentanamide
A solution of 6. Og (22.6 mmol) of N-CBZ-L-isoleucine in 45 mL of anhydrous DMF was cooled to 0°C and charged with 4.0g (29.5 mmol) of HOBT and 4.3g (22.6 mmol) of EDC. The ice bath was removed after 20 minutes and stirring was continued for an additional 40 minutes. The reaction solution was then charged with a solution of 7.4g (19.7 mmol) of 2R-hydroxy-3-[ (phenylsulfonyl) (2-methylpropyl) amino] -IS- (phenylmethyl) propylamine and 2.3g (22.6 mmol) of 4-methylmorpholine in 25 mL of anhydrous DMF and stirred for 18 hours. The solvents were removed in vacuo and the residue was partitioned between 300 mL of ethyl acetate and 120 mL of 5% potassium hydrogen sulfate solution. The layers were separated, and the organic layer was washed with 120 mL each of saturated sodium bicarbonate solution, water and brine, then dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo to afford 13g of crude material. The crude material was crystallized in ethanol. The solid was isolated by filtration, rinsed with one 50 mL portion of hexane, and air-dried to yield 10.3g (84%) of the desired product, m/e = 630 (M+Li) .
Part B: Preparation of N-[2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl] -2S-amino-3S-methylpentanamide
A Fischer-Porter bottle equipped with a magnetic stir bar was charged with 10.2g (16.4 mmol) of the product from Part A and 75 mL of tetrahydrofuran (THF) . The solution was hydrogenated in the presence of 4g of 10% palladium-on-carbon catalyst (50% water by weight) under 50 psig of hydrogen for 3 hours at room temperature. The catalyst was removed by filtration, and the solvents removed under reduced pressure. The residue was dissolved in 300 mL of ethyl acetate and washed with 120 mL each of saturated sodium bicarbonate solution and brine, then dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to afford 7.4g of the desired product, m/e = 490 (M+H) .
Part C: Preparation of N-[2R-hydroxy-3- [ (phenylsulfonyl) (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-l- yl) acetyl] amino] -3S-methylpentanamide
A solution of 3.4g (20.2 mmol) of pyrrolidineacetic acid hydrochloride in 45 mL of anhydrous DMF was cooled to 0°C and charged with 3. lg (22.7 mmol) of HOBT and 3.3g (17.4 mmol) of EDC. The ice bath was removed after 20 minutes and stirring was continued for an additional 40 minutes. The reaction solution was then charged with a solution of 7.4g (16.5 mmol) of the amine from Part B and 4. lg (40.4 mmol) of 4-methylmorpholine in 25 mL of anhydrous DMF and stirred for 17 hours. The solvents were removed in vacuo and the residue was partitioned between 300 mL of ethyl acetate and 120 mL of 5% potassium hydrogen sulfate solution. The layers were separated, and the organic layer was washed with 120 mL each of saturated sodium bicarbonate solution, water and brine, then dried over anhydrous magnesium sulfate, filtered and concentrated to afford 8.7g of crude material. The coupling reaction was run again using 2.3g (13.9 mmol) of pyrrolidinylacetic acid hydrochloride, 2.1 grams (15.5 mmol) of HOBT, 2.3 grams (12.0 mmol) of EDC, 2.8 grams (27.3 mmol) of 4- methylmorpholine, and the 8.7 grams of crude product in place of the amine from Part B. The reaction work-up was repeated and yielded 8.3 grams of crude product. Purification was accomplished using a Prep 2000 chromatograph on silica gel using 40-70% (5% methanol/95% ethyl acetate) /hexane and yielded 5.2 grams (57%) of the desired product as a white solid, m/e = 607 (M+Li) .
EXAMPLE 42
Preparation of N-f f2R-hydroxy-3-r (1.3-benzodioxol-5- γl) sulfonyl] (2-methylpropyl) amino1-1S- (phenylmethy1) propyl1 -2S- f f (pyrrolidin-l-y1) acetyl1 amino] -3- (methylsulfonyl) propaneamide'hvdrochloride salt
Figure imgf000130_0001
Part A: Preparation of N-[ [2R-hydroxy-3-[ (1,3- benzodioxol-δ-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) propyl] -2S-[ [ (1, 1-dimethylethoxy) carbonyl] amino] -3- (methylthio) propaneamide
Figure imgf000130_0002
N-t-Boc-S-methyl-(L) -cysteine (2.80 g, 11.9 mmol), 1- Hydroxybenzotriazole hydrate (1.92 g, 12.5 mmol), and 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (2.27 g, 11.9 mmol) were mixed in N,N- dimethyIformamide (30.0 mL) at 0°C for 10 min. N- Methylmorpholine (3.03 g, 33.0 mmol) was added and the solution stirred an additional 10 min at 0°C. 2R- Hydroxy-3-[ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propylamine (5.00 g, 11.9 mmol) was added and the solution was warmed to room temperature and stirred for 2 hours. The reaction mixture was poured into ethyl acetate (500 mL) and washed with 10% aqueous hydrochloric acid (3 X 100 mL) , saturated aqueous sodium bicarbonate (3 X 100 mL) and brine (2 X 100 mL) . The organic layer was dried over sodium sulfate and percolated through a bed of silica gel (50 g) . The desired product (7.13 g, 11.19 mmol, 93% yield) was obtained as a white solid by removal of the solvent at reduced pressure; m/e calcd 637; found (M + Li) 644.
Part B: Preparation of N-[ [2R-hydroxy-3-[ (1,3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S- [ [ (1, 1-dimethylethoxy) carbonyl] amino] -3- (methylsulfonyl) propaneamide
Figure imgf000131_0001
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl]-2S-[ [ (1, 1- dimethylethoxy) carbonyl] amino] -3- (methylthio) propaneamide (7.10 g, 11.1 mmol) was dissolved in methanol (150 mL) . A solution of oxone® (20.8 g, 33.9 mmol) in water (150 mL) was added dropwise to the solution at room temperature over 1.5 hours. The solution became cloudy and a precipitate formed during the addition. The reaction was stirred for an additional 1 hour and tetrahydrofuran (200 mL) was added. After an additional 1 hour of mixing the solution was poured into ethyl acetate (1000 mL) and washed with water (3 x 200 mL) followed by brine (2 x 300 ml) . The organic layer was dried over anhydrous sodium sulfate and solvent removed at reduced pressure. The desired product (5.75 g, 8.86 mmol, 79% yield) was obtained as an off white solid; m/e calcd 669; found (M+H) 670.
Part C: Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenyl ethyl) propyl] -2S-amino-3- (methylsulfonyl) propaneamide'hydrochloride salt
Figure imgf000132_0001
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropy1) amino] -IS- (phenylmethyl) propyl] -2S- [ [ (1, 1- dimethylethoxy) carbonyl] amino] -3- (methylsulfonyl) propaneamide (5.5 g, 8.20 mmol) was dissolved in dichloromethane (100 mL) at room temperature. Anhydrous hydrochloric acid was bubbled through the solution for 15 min. The solution was stirred at room temperature for 2 hours and the solvent was removed at reduced pressure. The desired product (4.91 g, 8.10 mmol, 99 % yield) was obtained as a white solid; m/e calcd 569; found (M+Li) 576.
Part D: Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S- [ (chloroacetyl) amino] -3- (methylsulfonyl) propaneamide
Figure imgf000133_0001
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -is- (phenylmethyl) propyl] -2S-amino-3- (methylsulfonyl)propaneamide»hydrochloride salt (4.00 g, 6.59 mmole) was mixed at room temperature in acetonitrile (40 mL) . Triethylamine (2.10 g, 21.0 mmol) and chloroacetic anhydride (1.12 g, 6.59 mmol) were added. The solution was stirred at room temperature for 16 hours and poured into ethyl acetate (250 mL) . The solution was washed with 10% aqueous acetic acid (2 x 100 mL) , saturated aqueous sodium bicarbonate (2 x 100 mL) , and brine (2 x 100 mL) . The organic layer was dried over anhydrous sodium sulfate and solvent removed at reduced pressure. The product (1.20 g, 1.85 mmol, 28% yield) was obtained as a white solid by crystallization from ethyl acetate and hexanes; m/e calcd 645; found (M+Li) 652.
Part E: Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -3- (methylsulfonyl) propaneamide
Figure imgf000133_0002
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl)propyl] -2S- [ (chloroacetyl) amino] -3- (methylsulfonyl) propaneamide (1.2 g, 1.85 mmol) and pyrrolidine (0.79 g, 11.14 mmol) were mixed together in tetrahydrofuran (12.0 mL) at room temperature for 5 hours. The solvent was removed at reduced pressure and the residue was taken up in ethyl acetate (150 mL) . The solution was washed with saturated sodium bicarbonate (l x 100 mL) , saturated ammonium chloride (1 x 100 mL) , and finally saturated sodium bicarbonate (1 x 100 mL) . The organic layer was dried over anhydrous sodium sulfate and solvent removed at reduced pressure. A white foam was collected. The product (653 mg, 0.95 mmol, 52% yield) was obtained as a white solid; m/e calcd 680; found (M+Li) 687.
Part F: Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -3- (methylsulfonyl) propaneamide»hydrochloride salt
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ [ (pyrrolidin-l-yl) acetyl] amino] -3- (methylsulfonyl) propaneamide (300 mg, 0.44 mmol) was dissolved in acetonitrile (15.0 mL) . Concentrated aqueous hydrochloric acid (100 uL, 1.2 mmol) was added and the solvent was removed at reduced pressure. The residue was taken up in acetonitrile (30 mL) and solvent removed at reduced pressure. The residue was taken up in water (10 mL) and solvent removed at reduced pressure. The aqueous solvent treatment was repeated two more times. The resulting white foam was dried for 16 hours at reduced pressure (0.5 mm Hg) at room temperature and the further dried over phosphorous pentoxide at reduced pressure (0.5 mm Hg) for 72 hours. The hydrochloric acid salt (313 mg, 0.44 mmol) was obtained as a white foam; m/e calcd 680; found (M+Li) 687.
EXAMPLE 43
Preparation of N-f r2R-hγdroxy-3- (1, 3-benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyI) ropyl] -2S- f (pyrrolidin-1- γl) acetyl1 amino1 -3-methyl-3-
(methylsulfonyl) butaneamide'hydrochloride salt
Figure imgf000135_0001
Part A: Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S-
(phenylmethy1)propyl] -2S-[ [ (1, l-dimethylethoxy) carbonyl] amino] -3-methy1-3- (methylthio) butaneamide
Figure imgf000135_0002
The N-t-boc-S-methyl-L-penicillamine dicyclohexylamine salt (4.00g, 9.00 mmol), 1-Hydroxybenzotriazole hydrate (1.69 g, 11.00 mmol), and l- (3-dimethylaminopropyl) -3- ethylcarbodiimide hydrochloride (1.71 g, 9.00 mmol) were mixed in dimethylformamide (60.0 mL) at room temperature. The heterogeneous mixture was stirred for 1 hour and 2R-hydroxy-3-[ [ (1, 3-benzodioxol-5- yl) sulfonyl] (2-methylpropyl) amino] -1S-
(phenylmethyl) propylamine (3.78 g, 9.00 mmol) was added and the heterogenous mixture was stirred for 16 hours. The solution was poured into ethyl acetate (600 mL) and washed with 10% aqueous acetic acid (2 x 300 mL) , saturated aqueous sodium bicarbonate (2 x 300 mL) and brine (300 mL) . The solution was dried over sodium sulfate and the solvent was removed in vacuo . The desired product was purified by flash chromatography (0- 80% ethyl acetate/hexanes on silica gel) . The product (5.21 g, 7.83 mmol, 87% yield) was obtained as a white foam; m/e calcd 665; found (M+Li) 672.
Part B: Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-[ [ (1, l-dimethylethoxy) carbonyl] amino] -3-methy1-3- (methylsulfonyl) butaneamide
Figure imgf000136_0001
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-[ [ (1,1- dimethylethoxy) carbonyl] amino] -3-methyl-3- (methylthio) butaneamide (5.01 g, 7.53 mmol) was dissolved in tetrahydrofuran (250 mL) . A solution of oxone® (13.8 g, 22.6 mmol) in water (250 mL) was added dropwise to the solution at room temperature over 2 hours. The solution became cloudy and a precipitate formed during the addition. The solution was poured into ethyl acetate (500 mL) and washed with water (3 x 200 mL) followed by brine (2 x 300 mL) . The organic layer was dried over anhydrous sodium sulfate and solvent removed in vacuo . The product (4.72 g, 6.77 mmol, 89% yield) was obtained as a white foam; m/e calcd 697; found (M+Li) 704.
Part C: Preparation of N-[ [2R-hydroxy-3-[ (1, 3- benzodioxol-5-y1) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-amino-3-methyl-3- (methylsulfonyl) butaneamide»hydrochloride salt
Figure imgf000137_0001
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -is- (phenylmethyl) propyl] -2S-[ [ (1,1- dimethylethoxy) carbonyl] amino] -3-methyl-3- (methylsulfonyl) butaneamide (4.51 g, 6.46 mmol) was dissolved in dichloromethane (200 mL) at room temperature. Anhydrous hydrochloric acid was bubbled through the solution for 30 min. The solution was stirred at room temperature for 1 hour and the solvent was removed in vacuo . The product (4.02 g, 6.35 mmol,
99% yield) was obtained as a white solid; m/e calcd 697; found (M+Li) 704.
Part D: Preparation of N-[ [2R-hydroxy-3- [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino]-lS-
(phenylmethy1) propyl ] -2S- [ (chloroacetyl) amino] -3-methy1- 3- (methylsulfonyl) butaneamide
Figure imgf000138_0001
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-amino-3- methyl-3- (methylsulfonyl) butaneamide»hydrochloride salt (3.90 g, 6.15 mmole) was mixed at room temperature in acetonitrile (40 mL) . Triethylamine (1.86 g, 18.45 mmol) and chloroacetic anhydride (1.05 g, 6.15 mmol) were added. The solution was stirred at room temperature for 16 hours and poured into ethyl acetate (250 mL) . The solution was washed with 10% aqueous acetic acid (2 x 100 mL) , saturated aqueous sodium bicarbonate (2 x 100 mL) , and brine (2 x 100 mL) . The organic layer was dried over anhydrous sodium sulfate and solvent was removed in vacuo . A yellow oil (4.3 g) was obtained and purified by flash chromatography
(silica gel, 50-75% ethyl acetates in hexanes. The product (2.15 g, 3,18 mmol, 52% Yield) was obtained as a white foam; m/e calcd 674; found (M+Li) 681.
Part E: Preparation of N-[ [2R-hydroxy-3-[ (1,3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl]-2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -3-methy1-3-(methylsulfonyl) butaneamide
Figure imgf000138_0002
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ (chloroacetyl) amino] -3-methyl-3- (methylsulfonyl) butaneamide (2.06 g, 3.05 mmol) and pyrrolidine (2.16 g, 30.5 mmol) were mixed together in tetrahydrofuran (30.0 mL) at room temperature for 3 hours. The solvent was removed at reduced pressure. A light yellow oil was obtained which was dissolved in methanol (20 mL) and the solvent was removed in vacuo . The product (2.09 g, 2.95 mmol, 96% yield) was obtained as a yellow foam; m/e calcd 708; found (M+H) 709.
Part F: Preparation of N-[ [2R-hydroxy-3-[ (1,3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino]-lS- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -3-methyl-3- (methylsulfonyl) butaneamide»hydrochloride salt
N-[ [2R-hydroxy-3-[ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-
[ [ (pyrrolidin-l-yl) acetyl] amino] -3-methyl-3- (methyl sulfonyl) butaneamide (300 mg, 0.42 mmol) was dissolved in dichloromethane (10 mL) . Anhydrous hydrochloric acid was added for 1 min and the dichloromethane solution was reduced to a volume of about 2 mL. The solution was added dropwise to hexanes (50 mL) and the product precipitated. The desired salt (295 mg, 0.40 mmol) was collected in a buchner funnel by vacuum filtration as a yellow solid; m/e calcd 708; found (M+H) 709.
EXAMPLE 44
Preparation of N- r2R-hydroxy-3- f f (1 , 1-dimethylethoxy) carbonyll (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-r f ( yrrolidin-l- yl) acetyl] amino] -3 , 3-dimethylbutanamide
Figure imgf000140_0001
Part A: Preparation of N-[(l,l- dimethylethoxy1) carbonyl] -N- [ 2-methylpropyl] -3S- [N1-
(phenylmethoxycarbonyl) amino] -2R-hydroxy-4- phenylbutylamine
Figure imgf000140_0002
A solution of N- [33-tN1- (benzyloxycarbonyl) amino] -2R- hydroxy-4-phenylbutyl]-N-(2-methylpropyl) amine (18.5 g, 50 mmol), BOC-ON (12.35 g, 50 mmol) and triethylamine (7 mL) in tetrahydrofuran (400 mL) was stirred at room temperature for 18 hours and then concentrated in vacuo . The residue was dissolved in dichloromethane (1 L) and washed with sodium hydroxide (5 %( 2 X 200 mL) and brine, dried (MgS04) and then concentrated in vacuo to afford 23.5 g (quantitative yield) of the pure desired product.
Part B: Preparation of N- [2R-hydroxy-3- [ [ (1, 1- dimethylethoxy) carbonyl] (2-methylpropyl) amino]-lS- (phenylmethyl) propyl] -2S- [ (phenylmethoxycarbonyl) amino] - 3 , 3-dimethylbutanamide
Figure imgf000141_0001
N-[ (1, l-dimethylethoxy1) carbonyl] -N-[2-methylpropyl] -3S- [N1- (phenyImethoxycarbony1) amino] -2R-hydroxy-4- phenylbutylamine in ethanol was hydrogenated at 45 psig of hydrogen in the presence of 5% pd(C) catalyst to yield N-[ (1, l-dimethylethoxy1) carbonyl]-N-[2- methylpropyl] -3S- [N1- (phenylmethoxycarbonyl) amino] -2R- hydroxy-4-phenylbutylamine. Following standard workup, the crude amine (12.24 g, 36.42 mmol) was added to a mixture of N-carbobenzyloxycarbonyl-L-tert-leucine (9.67 g, 36.42 mmol), HOBT (4.92 g, 36.42 mmol) and EDC (6.98 g, 36.42 mmol) in DMF (300 mL) after the mixture was stirred at room temperature for 1 hour. The mixture was stirring for an additional 18 hours. The DMF was removed in vacuo , the residue was dissolved in dichloromethane (500 mL) , washed with sodium hydroxide (5%, 2 X 200 mL) and brine (200 mL) , dried and concentrated to afford 21 g (quantitative) of the desired product.
Part C: Preparation of N-[2R-hydroxy-3-[ [ (1,1- dimethylethoxy) carbonyl] (2-methylpropyl) amino] -1S- (phenylmethy1) ropyl] -2S-amino-3 , 3-dimethylbutanamide
Figure imgf000141_0002
N-[2R-hydroxy-3-[ [ (1, l-dimethylethoxy) carbonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ (phenylmethoxycarbonyl) amino] -3 , 3-dimethylbutanamide (20 g, 34.29 mmol) in methanol (250 mL) was hydrogenated at room temperature in the presence of Pd/C (10%, 5 g) . The catalyst was filtered off and the filtrate was concentrated to afford 13.8 g (90%) of the pure desired product.
Part D: Preparation of N-[2R-hydroxy-3-[ [ (1,1- dimethylethoxy) carbonyl] (2-methylpropyl-amino] -1S- (phenylmethyl) propyl] -2S- [ (chloroacetyl) amino] -3,3- dimethylbutanamide
N^O^
Figure imgf000142_0001
To N-[2R-hydroxy-3-[ [ (1,1-dimethylethoxy) carbonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-amino- 3, 3-dimethylbutanamide (12.45 g, 27.70 mmol) in dichloromethane (200 mL) was added chloroacetic anhydride (5.21 g, 30.48 mmol) and the reaction mixture was stirred for 18 hours. The reaction mixture was washed with citric acid (5%, 100 mL) , sodium bicarbonate (saturated, 100 mL) and brine, dried (MgS04) and concentrated to afford 12.0 g (82%) of the pure desired product.
Part E: Preparation of N-[2R-hydroxy-3-[ [ (1, 1- dimethylethoxy) carbonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-l-yl) acetyl] amino] -3 , 3-dimethylbutanamide A mixture of N-[2R-hydroxy-3-[ [ (1, l-dimethylethoxy) carbonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-[ (chloroacetyl) amino] -3 , 3- dimethylbutanamide (10.42 g, 19.82 mmol) in tetrahydrofuran (100 mL) was cooled to 0°C, and pyrrolidine (7.1 g, 100 mmol) was added. The reaction mixture was stirred for 18 hours. The reaction mixture was concentrated and the residue was dissolved in ethyl acetate (500 mL) , washed with sodium bicarbonate (saturated, 200 mL) and brine (200 mL) , dried (MgS04) and concentrated to afford 11.0 g (quantitative) of the desired product.
EXAMPLE 45
Preparation of N-f2R-hydroxy-3-f fphenylsulfonyl] (2- methylpropyl) amino1 -IS- (phenylmethyl) propyl1 -2S-
[ [ (pyrrolidin-1-yl) acetyl] amino] -3.3-dimethylbutanamide
Figure imgf000143_0001
N-[2R-hydroxy-3-[ [ (1, l-dimethylethoxy) carbonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-
[ [ (pyrrolidin-l-yl) acetyl] amino] -3 , 3-dimethylbutanamide (2 g, 3.57 mmol) in Dioxane/HCl (4N, 10 mL) was stirred for 2 hours at room temperature. The solvent was removed and the residue was dried in vacuo . The residue was stirred in ethyl acetate (50 mL) then benzenesulfonyl chloride (0.692 g, 3.57 mmol) was added followed by triethylamine (1.587 g, 15.71 mmol) and the mixture was stirred for 18 hours at room temperature. The reaction mixture was diluted with ethyl acetate (100 mL) , washed with saturated sodium bicarbonate (saturated, 100 mL) and brine (100 mL) , dried (MgS04) , and concentrated. The residue was chromatographed in ethyl acetate to afford 5.0 g (47%) of the desired product as a white powder; m/e=601 (M+H) .
EXAMPLE 46
Preparation of N- [2R-hydroxy-3-f r (4-methoxyphenyl) sulfonyl] (2-methylpropyl) amino] -1S-
( henylmethyl) propyl1 -2S-f [ (pyrrolidin-l- yl) acetyl1amino]-3 , 3-dimethylbutanamide
Figure imgf000144_0001
N-[2R-hydroxy-3-[ [ (1, l-dimethylethoxy) carbonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ [ (pyrrolidin-l-yl) acetyl] amino] -3, 3-dimethylbutanamide (2 g, 3.57 mmol) in Dioxane/HCl (4N, 10 mL) was stirred for 2 hours at room temperature. The solvent was removed and the residue was dried in vacuo . The residue was stirred in ethyl acetate (50 mL) then 4- methoxybenzene sulfonyl chloride (0.737 g, 3.57 mmol) was added followed by triethylamine (1.587 g, 15.71 mmol) and the mixture was stirred for 18 hours at room temperature. The reaction mixture was diluted with ethyl acetate (100 mL) , washed with saturated sodium bicarbonate (saturated, 100 mL) and brine (100 mL) , dried (MgS04) , and concentrated. The residue was chromatographed in ethyl acetate to afford 1.0 g (44%) of the desired product as a white powder; m/e=631 (M+H) . EXAMPLE 47
Preparation of N-r2R-hydroxy-3-f [ (2.3-dihydrobenzofuran- 5-y1) sulfonyl1 (2-methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-[ f (pyrrolidino) acetyl] amino1-3 ,3- dimethylbutanamide
Figure imgf000145_0001
N-[2R-hydroxy-3-[ [ (1, l-dimethylethoxy) carbonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ C (pyrrolidin-l-yl) acetyl] amino] -3, 3-dimethylbutanamide (2 g, 3.57 mmol) in Dioxane/HCl (4N, 10 mL) was stirred for 2 hours at room temperature. The solvent was removed and the residue was dried in vacuo . The residue was stirred in ethyl acetate (50 mL) then 2,3-dihydro benzofuran-5-ylsulfonyl chloride (0.737 g, 3.57 mmol) was added followed by triethylamine (1.587 g, 15.71 mmol) and the mixture was stirred for 18 hours at room temperature. The reaction mixture was diluted with ethyl acetate (100 mL) , washed with saturated sodium bicarbonate (saturated, 100 mL) and brine (100 mL) , dried (MgS04) , and concentrated. The residue was chromatographed in ethyl acetate to afford the desired product as a white powder. EXAMPLE 48
Preparation of 2R-hydroχy-3-f f (l,4-benzodioxan-6-yl) sulfonyll (2-methylpropyl) aminol -IS- (phenylmethyl) propyl carbamic acid phenylmethyl ester
Figure imgf000146_0001
To a solution of the N-[3S-[ (phenylmethoxycarbonyl) amino]-2R-hydroxy-4-phenyIbutyl] -N- (2-methylpropyl) amine (0.5 g, 1.35 mmol) in CHC12 (5.0 mL) containing Et3N (0.35 mL, 2.5 mmol) was added 1, 4-benzodioxan-6-sulfonyl chloride (0.34 g, 1.45 mmol) and stirred at 0°C for 30 min. After stirring at room temperature for 1 hour, the reaction mixture was diluted with CHC12 (20 mL) , washed with cold IN HCl (3 x 20 mL) , water (2 x 20 mL) , satd. NaHC03 (2 x 20 mL) and water (3 x 20 mL) , dried (Na2S0 ) and concentrated under reduced pressure. The resulting residue was purified by flash chromatography using 35% EtOAc in hexane to give the desired product as a white amorphous solid which crystallized from MeOH as a white powder (0.65g. 84% yield): m.p. 82-84°C, HRMS-FAB : calcd for C30H37N2O7S 569.2321 (MH+) , found 569.2323. EXAMPLE 49
Figure imgf000147_0001
Preparation of 2S-f f (pyrrolidino) acetyl] amino] -N-f2R- hydroxy-3-f f (1, 3-benzodioxol-5-γl) sulfonyl] (2- methylpropyl) aminol -IS- (phenylmethyl) propyl1-3- methylpent-4-vnamide
Part A . Preparation of 2S-[ [ (1, l-dimethylethoxy) carbonyl] amino] -N- [2R-hydroxy-3- [[(1,3-benzodioxol-5- yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl]pent-4-ynamide
Figure imgf000147_0002
To a cooled solution of N-t-Boc-L-propargyl glycine (5.0 g, 23.4 mmol) and 4.7g (1.5 equiv.) of N-hydroxy benzotriazole in 40 mL of N,N-dimethylformamide was added 4.6g (23.4 mmol) of EDC and stirred at 0°C for one hour. To this was added a solution of 12.10g (23.4 mmol) of 2R-hydroxy-3-[ [ (1, 3-benzodioxol-5- yl) sulfonyl] (2-methyl propyl) amino] -1S-
(phenylmethyl) propylamine in 6 mL of N,N- dimethylformamide and the solution stirred for 16 hours.
The solvent was removed by rotory evaporation, replaced with ethyl acetate, and washed with saturated sodium bicarbonate, 5% citric acid and brine. The organics were dried over magnesium sulfate, filtered and concentrated to yield 13.3 grams of crude product, which was crystallized from diethyl ether: ethyl acetate to yield 6.9g of 2S-[ [ (1, l-dimethylethoxy) carbonyl] amino] -N-[2R- hydroxy-3-[[ (1, 3-benzodioxol-5-yl) sulfonyl] (2-methyl propyl) amino] -IS- (phenylmethyl) propyl]pent-4-ynamide.
Part B. Preparation of 2S-amino-N-[2R-hydroxy-3-[ [ (1, 3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl]pent-4-ynamide
Figure imgf000148_0001
5.0g (8.12 mmol) of the product from Part A. was dissolved in 20 mL of 4N HCl in dioxane and stirred for 30 minutes. The precipitated product was stripped two times from diethyl ether and this crude hydrochloride salt was used in Part C.
Part C. Preparation of 2S-[ (chloroacetyl) amino]-N-[2R- hydroxy-3-[ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2-methyl propyl) amino] -IS- (phenylmethyl) propyl]pent-4-ynamide
Figure imgf000148_0002
4.4g (8.12 mmol) of amine hydrochloride from Part B was dissolved in 60 mL of methylene chloride and to this was added 3. Og (24 mmol) of N,N-diisopropylethyl amine, followed by 1.38g (8.1 mmol) of chloroacetic anhydride. The solution was stirred at room temperature overnight. The contents were concentrated on a rotory evaporator and the residue was partitioned between ethyl acetate and water. The organic layer was washed with 5% citric acid and then saturated sodium bicarbonate and brine. The organics were dried over magnesium sulfate filtered and concentrated to yield 4.3g of crude*product. Recrystallization from ethyl acetate hexane yielded 3.6 g (75% yield) of 2S-[ (chloroacetyl) amino]-N-[2R-hydroxy- 3-[[ (l,3-benzodioxol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl]pent-4- ynamide as a white solid.
Part D. Preparation of 2S-[ [ (pyrrolidino) acetyl] amino] - N-[2R-hydroxy-3-[ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2- methyl propyl) amino] -IS- (phenylmethyl) propyl]pent-4- ynamide
Figure imgf000149_0001
2S-[ (Chloroacetyl) amino]-N-[2R-hydroxy-3-[ [ (1,3- benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl]pent-4-ynamide (3.5 g, 6.0 mmol) was dissolved in 30 mL of tetrahydofuran and to this was added 2.3 g (5 equiv.) of pyrrolidine and the reaction stirred for 1.5 hours. The solvents were removed by rotory evaporation and replaced with ethyl acetate. The organics were washed successively with saturated sodium bicarbonate, and brine, dried over magnesium sulfate filtered and concentrated to yield 3.4 grams of crude product. Purification by crystallization from diethyl ether yielded 3.0 grams of 2S-[ [ (pyrrolidino) acetyl] amino] -N-[2R-hydroxy-3-[ [ (1, 3-benzodioxol-5-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl)propyl]pent-4- ynamide as a white solid.
EXAMPLE 50
Preparation of 5-chlorosulfonyl-2-carbomethoxγamino- benzimidazole
Figure imgf000150_0001
A solution of 2-carbomethoxyamino-benzimidazole (5.0 g, 0.026 mole) in chlorosulfonic acid (35.00 L) was stirred at 0°C for 30 minutes and at room temperature for 3 hours. The resulting dark colored reaction mixture was poured into an ice-water mixture (200 mL) , and stirred at room temperature for 30 minutes. The resulting precipitate was filtered and washed with cold water (500 mL) . The solid was dried overnight under high vacuum in a desiccator over NaOH pellets to give 5- chlorosulfonyl-2-carbomethoxyamino-benzimidazole (5.9 g, 78%) as a gray powder. 1H NMR (DMSO-d6) d: 3.89 (s,
3H) , 7.55 (d, J = 8.4 Hz, 1H) , 7.65 (d, J" = 8.4 Hz, 1H) , 7.88 (s, 1H) . (German Patent DE 3826036)
EXAMPLE 51
Preparation of N-f2R-hvdroxy-3-rN:1—r (2- carbomethoxyamino-benzimidazol-5-yl) sulfonyll-N1- (2- methylpropyl) aminol -IS- (phenylmethyl) propyl]carbamic acid phenylmethyl ester
Figure imgf000151_0001
To a cold solution of N-[3S-[ (phenyImethoxycarbony1) amino] -2R-hydroxy-4-phenyIbutyl] -N- (2-methylpropyl) amine (5.0 g, 13.5 mmol) in dichloromethane (70 mL) was added triethylamine (5.95 g, 54.0 mmol) followed by the addition of 5-chlorosulfonyl-2-carbomethoxyamino- benzimidazole (4.29 g, 14.85 mmol) in small portions as a solid. The reaction mixture was stirred at 0°C for 30 minutes and at room temperature for 2.5 hours when reaction of the amino alcohol was complete. The mixture was cooled and filtered, and the filtrate was concentrated. The resulting residue was dissolved in EtOAc (200mL) , washed successively with cold 5% citric acid (3 x 50 mL) , saturated aqueous sodium bicarbonate (3 x 50 mL) and water (3 x 100 mL) , then dried (Na2S04) , concentrated and dried under vacuum. The residue was triturated with methanol, cooled, filtered, washed with MeOH-EtOAc (1:1, v/v) and dried in a desiccator to give pure N-[2R-hydroxy-3-[ [ (2-carbomethoxyamino- benzimidazol-5-yl) sulfonyl] (2-methylpropyl) -amino] -1S- (phenylmethyl) propyl] carbamic acid phenylmethyl ester (6.02 g, 72 %) as a light brown powder: FABMS: m/z = 630 (M+Li); HRMS: calcd. for C31H38N507S (M+H) 624.2492, found 624.2488. EXAMPLE 52
Preparation of 2R-hvdroxy-3-f f (2-amino-benzimidazol-5- yl) sulfonyl! (2-methyl-propyl) amino] -IS- (phenylmethyl) propylamine
Figure imgf000152_0001
A solution of N-[2R-hydroxy-3-[ [ (2-carbomethoxyamino- benzimidazol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] carbamic acid phenylmethyl ester (0.36 g, 0.58 mmol) in 2.5 N methanolic KOH (2.00 mL) was heated at 70°C under a nitrogen atmosphere for 3 hours. The reaction mixture was diluted with water (10 mL) and extracted with EtOAc (3 x 15 mL) . The combined organic extracts were washed with brine, dried (Na2S04) and concentrated. The resulting residue was purified by reverse-phase HPLC using a 10-90% CH3CN/H0 gradient (30 min) at a flow rate of 70 mL/min. The appropriate fractions were combined and freeze dried to give pure 2R-hydroxy-3-[ [ (2-amino-benzimidazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenyl-methyl) propylamine (0.22 g, 58%) as a white powder: FAB-MS m/z = 432 (M+H); HRMS: calcd. for C21H30N503S (M+H) 432.2069, found 432.2071. EXAMPLE 53
Preparation of N-r2R-hvdroxy-3-f f (2-amino-benzimidazol- 5-yl) sulfonyl1 (2-methylpropyl) -aminol -IS- (phenylmethyl) propyl] carbamic acid phenylmethyl ester
Figure imgf000153_0001
To a solution of 2R-hydroxy-3-[ [ (2-amino-benzimidazol-5- yl) sulfonyl] (2-methyl-propyl) amino] -IS- (phenylmethyl) propylamine (0.22 g, 0.33 mmol) in THF (3.00 mL) , triethylamine (0.11 g, l.l mmol) and benzyloxycarbonyl succinimide (0.09 g, 0.36 mmol) were added, and the reaction mixture was stirred at room temperature for 16 hours. The solution was concentrated, and the residue was partitioned between EtOAc (15 mL) and saturated aqueous sodium bicarbonate. The organic phase was washed with brine, dried (Na2S0 ) , and concentrated. The resulting residue was purified by reverse-phase HPLC using a 10-90% CH3CN/H20 gradient (30 min) at a flow rate of 70 mL/min. The appropriate fractions were combined and freeze dried to give pure N-[2R-hydroxy-3- [ [ (2-amino-benzimidazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] carbamic acid phenylmethyl ester (0.12 g, 61%) as a white powder: FAB-MS m/z = 566 (M+H); HRMS: calcd. for C29H36N505S 566.2437 (M+H), found 566.2434. EXAMPLE 54
Preparation of 2R-hydroxγ-3-f f (2-carbomethoxyamino- benzimidazol-5-yl) sulfonyl] (2-methylpropyl) aminol -1S- (phenylmethyl) propylamine
Figure imgf000154_0001
A solution of N-[2R-hydroxy-3-[ [ (2-carbomethoxyamino- benzimidazole-5-yl) sulfonyl] (2-methylpropyl) -amino] -1S- (phenylmethyl) propyl] carbamic acid phenylmethyl ester (2.5 g, 0.4 mmol) in MeOH (10 L) and THF (50 mL) was hydrogenated in the presence of 10% Pd/C (1.2 g) at room temperature at 60 psi for 16 hours. The catalyst was removed by filtration, and the filtrate was concentrated under reduced pressure. The resulting residue was triturated with ether and" filtered. The solid substance thus obtained was washed with ether and dried in vacuo to afford pure 2R-hydroxy-3-[ [ (2-carbomethoxyamino- benzimidazol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propylamine (1.5 g, 77%) as an off white powder: Rt =12.8 min; FAB-MS m/z = 490 (M+H); HRMS: calcd. for C23H32N505S 490.2124 (M+H), found 490.2142.
EXAMPLE 55
Preparation of N-f2R-hvdroxy-3-[ [ (2-carbomethoxyamino- benzimidazol-5-yl) sulfonyl] (2-methylpropyl) aminol -1S- (phenylmethyl) propyl] -2S-amino-3 , 3-dimethylbutanamide
Figure imgf000155_0001
Part A: Preparation of N-[2R-hydroxy-3-[N1-[ (2- carbomethoxyamino-benzimidazol-5-yl) sulfonylY-N1- (2- iethylpropyl) amino] -IS- (phenylmethyl) propyl-2S-
[ (phenylmethoxy-carbonyl) amino] -3 , 3-dimethylbutanamide
Figure imgf000155_0002
To a solution of N-carbobenzyloxycarbonyl-L-tert- leucine (0.65 g, 2.45 mmol) in DMF (10 mL) was added HOBt (0.5 g, 3.22 mmol) and EDC (0.49 g, 2.55 mmol), and the resulting mixture was stirred at 0°C for 2 hours. Then a solution of 2R-hydroxy-3-[ [ (2- carbomethoxyamino-benzimidazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propylamine (1.2 g, 2.45 mmol) in DMF (4 mL) and N-methyl morpholine (0.74 g, 7.3 mmol) was added, and the mixture was stirred at room temperature for 16 hours. The DMF was then distilled away in vacuo , and the remaining residue was partitioned between cold IN aqueous HCl (100 mL) and EtOAc (200 mL) . The organic phase was washed successively with cold IN HCl (2 x 50 mL) , brine (2 x 50 mL) , 0.25 N NaOH (3 x 50 mL) , brine, dried (Na2S04) and concentrated in vacuo . The resulting residue was purified by silica gel flash column chromatograthy using EtOAc as the eluent to afford 1.5 g (83%) of pure N-[2R-hydroxy-3-[ [ (2-carbomethoxyamino-benzimidazol-5- yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl-2S- [ (phenylmethoxy-carbonyl) amino] -3 , 3-dimethyl butanamide: Rt = 21.2 min; FAB-MS m/z = 737 (M+H), HRMS: calcd. for C37H49N608S 737.3333 (M+H), found 737.3334.
Part B: Preparation of N-[2R-hydroxy-3-[ [ (2- carbomethoxyamino-benzimidazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-amino- 3,3-dimethylbutanamide
A solution of N-[2R-hydroxy-3-[ [ (2-carbomethoxyamino- benzimidazol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl-2S- [ (phenylmethoxycarbonyl) amino]- 3, 3-dimethylbutanamide (4.0 g, 5.4 mmol) in MeOH (15 mL) and THF (65 mL) was hydrogenated in the presence of 10% Pd/C (2.0 g) at room temperature at 50 psi for 16 hours. The catalyst was removed by filtration, and the filtrate was concentrated under reduced pressure. The resulting residue was triturated with ether and filtered. The solid residue was washed with ether and dried in vacuo to afford N-[2R-hydroxy-3-[ [ (2-carbomethoxyamino- benzimidazol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-amino-3 , 3-dimethylbutanamide (2.9 g, 88%) as a pale yellow powder. A portion of the material was purified by reverse-phase HPLC using a 10- 90% CH3CN/H20 gradient (30 min) at a flow rate of 70 mL/min. The appropriate fractions were combined and freeze dried to give pure N-[2R-hydroxy-3-[ [ (2- carbomethoxyamino-benzimidazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl-2S-amino- 3, 3-dimethylbutanamide as a white powder: Rt = 13.9 min; FAB-MS m/z = 609 (M+Li), 603 (M+H); HRMS: calcd. for C29H43N606S 603.2965 (M+H), found 603.2972.
EXAMPLE 56
Preparation of N-r2R-hydroxy-3-f fN1-(2- carbomethoxyamino-benzimidazol-5-yl) sulfonyl] -N1-(2- methylpropyl) aminol -IS- (phenylmethyl) propyl! -2S- f f (pyrrolidin-l-yl) acetyl] aminol-3 ,3- dimethylbutanamide
Figure imgf000157_0001
Part A: Preparation of N-[2R-hydroxy-3-[ [ (2- carbomethoxy amino-benzimidazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl-2S- [ (chloroacetyl) amino] -3 , 3-dimethylbutanamide
Figure imgf000157_0002
A mixture of chloroacetic acid (0.32 g, 3.39 mmol), HOBt (0.78 g, 5.0 mmol), and EDC (0.65 g, 3.39 mmol) in DMF (5 mL) was stirred at 0°C for 1 hour, and was then added to a solution of N- [2R-hydroxy-3-[ [ (2- carbomethoxyamino-benzimidazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S-amino- 3, 3-dimethylbutanamide (2.0 g, 3.3 mmol) in DMF (5 mL) . The resulting mixture was stirred at 0°C for 2 hours, and at room temperature for 1 hour when the reaction was complete. The DMF was removed in vacuo . The resulting residue was dissolved in EtOAc (50 mL) and washed successively with saturated aqueous sodium bicarbonate (3 x 25 mL) , brine, dried (Na S0 ) , and concentrated under reduced pressure. The resulting material was crystallized from EtOAc to give 1.2 g (53%) of pure N-[2R-hydroxy-3- [ [ (2-carbomethoxy amino- benzimidazol-5-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S- [ (chloroacatyl) amino] -3,3- dimethylbutanamide as a shite powder: m.p. 253 °C
(decomp) ; Rt 18.1 min; FAB-MS m/z = 679*(M+H), HRMS: calcd. for C31H44N607SC1 679.2681 (M+H), found 679.2690.
Part B. Preparation of N-[2R-hydroxy-3-[ [ (2- carbomethoxyamino-benzimidazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ [ (pyrrolidin-l-yl) acetyl] amino] -3 , 3- dimethylbutanamide.
N-[2R-hydroxy-3-[ [ (2-carbomethoxyamino-benzimidazol-5- yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ (chloroacetyl) amino] -3 , 3-dimethylbutanamide (0.5 g, 0.74 mmol) was dissolved in THF (2.00 mL) , pyrrolidine (0.3 g, 4.2 mmol) was added, and the mixture was stirred at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure, and the residue was dried in vacuo . The resulting material was triturated with 10% EtOAc in ether and filtered. The solid was washed with ether and dried to give 0.42 g of crude product as a pale yellow powder. This was purified by reverse-phase HPLC using a 5)70% CH3CN/H20 gradient (30 min) at a flow rate of 70 mL/min. The appropriate fractions were combined and freeze dried to give pure N- [2R-hydroxy-3- [ [ (2- carbomethoxyamino-benzimidazol-5-yl) sulfonyl] (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [ [ (pyrrolidin-l-yl) acetyl] amino] -3 , 3- dimethylbutanamide (0.41 g, 77%) as a white powder: R = 14.8 min; FAB-MS m/z = 714 (M+H), HRMS: calcd. for C35H52N707S 714.3649 (M+H), found 714.3666.
EXAMPLE 57
Preparation of N-f 2R-hydroxy-3-f f (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl-2S-f f (pyrrolidin-l- yl) acetyl 1 amino1-3 ,3-dimethyl butanamide*dihvdrochloride
Figure imgf000159_0001
Part A: Preparation of N-[2R-hydroxy-3-[ (2- methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [[(pyrrolidin-l-yl) acetyl] amino] -3 ,3- dimethylbutanamide»dihydrochloride
Figure imgf000159_0002
A solution of hydrochloric acid in dioxane (4N, 10 mL) was added to N-[2R-hydroxy-3-[ [ (1, l-dimethylethoxy) - carbonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl] - S~ [ [ (pyrrolidin-l-yl) acetyl] amino] -3 , 3- dimethylbutanamide (2.80 g, 5.0 mmol), and the mixture was stirred for 2 hours at room temperature. The solvent was removed, and the residue was dried in vacuo to afford 2.60 g of the desired dihydrochloride product as a crystalline solid. Part B: Preparation of N-[2R-hydroxy-3- [ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-1- yl) acetyl] amino] -3 , 3-dimethylbutanamide»dihydrochloride
To a solution of 6-chlorosulfonylbenzothiazole in dichloromethane (100 mL) was added N-[2R-hydroxy-3-[ (2- methylpropyl) amino] -IS- (phenylmethyl) -propyl]-2S- [ [ (pyrrolidin-l-yl) acetyl] amino] -3,3- dimethylbutanamide»dihydrochloride (l.OO^g, 1.875 mmol) and neat triethylamine (3 mL) . After stirring at room temperature for 18 hours, the reaction mixture was diluted with dichloromethane (100 mL) , washed with saturated aqueous sodium bicarbonate (100 mL) , brine
(100 mL) , dried (MgS0 ) , filtered and concentrated. The resulting residue was purified by flash column chromatography on silica gel, eluting with 5% methanol in ethyl acetate to afford pure N-[2R-hydroxy-3- [ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -2S-[ [ (pyrrolidin-l- yl) acetyl] amino] -3 , 3-dimethyl butanamide (0.180 g, 15%) . This was converted to the dihydrochloride salt by concentrating an acetonitrile solution with IN HCl (2 mL) . The residue was then dried to afford the desired dihydrochloride salt: FAB-MS C33H47N505S2 : m/z = 657.
EXAMPLE 58
Preparation of N-r2R-hydroxy-3-f f (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propyl] -2S-f (chloroacetyl) amino] -3.3- dimethylbutanamide
Figure imgf000161_0001
Part A: Preparation of [2R-hydroxy-3-[ (4- a inophenylsulfonyl) (2-methylpropyl) amino] -1S- (phenylmethyl) propylcarbamic acid t-butyl ester
Figure imgf000161_0002
A mixture of [2R-hydroxy-3-[ (4-aminophenylsulfonyl) (2- methylpropyl) -amino] -IS- (phenylmethyl) propylamine 3.7 g (9.45 mmol) and BOC-ON (2.33 g, 9.45 mmol) and triethylamine (0.954 g, 9.45 mmol) in tetrahydrofuran (60 mL) was stirred for 16 h and concentrated in vacuo . The residue was dissolved in dichloromethane (200 mL) , washed with sodium hydroxide (IN, 100 mL) , citric acid (5%, 100 mL) , dried (MgS04), and concentrated to afford 1.18 g (94%) of the desired product as a white solid.
Part B: Preparation of [2R-Hydroxy-3-[ (2- aminobenzothiazole-6-sulfonyl) - (2-methylpropyl) amino] - IS- (phenylmethyl) propylcarbamic acid t-butyl ester
Figure imgf000162_0001
" cfr NH2
The [2R-hydroxy-3-[ (4-aminophenylsulfonyl) (2- methylpropyl) ]amino] -IS- (phenylmethyl) propylcarbamic acid t-butyl ester 1.12 g (2.279 mmol) was added to a well mixed powder of anhydrous copper sulfate (4.48 g) and potassium thiocyanate (5.60 g) followed *by dry methanol (35 mL) and the resulting black-brown suspension was heated at reflux for 2h. The reaction mixture turned gray. The reaction mixture was filtered and the filtrate was diluted with water (50 mL) and heated at reflux. Ethanol was added to the reaction mixture, cooled and filtered. The filtrate upon concentration afforded a residue which was chromatographed (ethyl acetate:methanol 90:10) to afford 0.80 g (78%) of the deprotected compound as a solid. This was directly reprotected via the following procedure; (2.25 g, 5.005 mmol) BOC-ON (1.24 g) , and triethylamine (0.505 g, 5.005 mmol) in tetrahydrofuran (20 mL) was stirred at room temperature for 18h. The reaction mixture was concentrated and the residue was dissolved in dichloromethane (200 mL) and was washed with sodium hydroxide (IN, 100 mL) , citric acid (5%, 100 mL) dried (Mg S04) and concentrated to afford a residue which was chromatographed (ethyl acetate:hexane 3:1) to afford 1.8 g (65%) of the desired product as a solid.
Part C: Preparation of [2R-hydroxy-3-[ (benzothiazole- 6-sulfonyl) (2-methylpropyl) amino] -IS- (phenylmethyl) propylcarbamic acid t-butyl ester
Figure imgf000163_0001
The product of part B above (1.80 g, 3.2755 mmol) was added to a solution of isoamylnitrite (0.88 mL) in dioxane (20 mL) and the mixture was heated at 85°C. After the cessation of evolution of nitrogen, the reaction mixture was concentrated and the residue was purified by chromatography (hexane: ethyl acetate 1:1) to afford 1.25 g (78%) of the desired product as a solid.
Part D: Preparation of [2R-hydroxy-3-[ (benzothiazole- 6-sulfonyl) (2-methylpropyl) amino] -IS- (phenylmethyl) propylamine»hydrochloride
Figure imgf000163_0002
The product of part C above was deprotected via the following procedure; (1.25 g, 2.3385 mmol) was added dioxane/HCl (4N, 10 mL) and was stirred at room temperature for 2h and concentrated. Excess HCl was chased with toluene to afford 1.0 g (quantitative yield) of the desired product as its HCl salt.
Part E: Preparation of N- [2R-hydroxy-3- [ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) -amino] IS- (phenylmethyl) propyl] -2S-[ [ (N- benzyloxy) carbonyl] amino] -3 , 3-dimethyl butanamide
Figure imgf000164_0001
A mixture of N-benzyloxycarbonyl-t-butylglycine (2.0 g, 7.538 mmol), HOBT (1.02 g, 7.55 mmol), and EDC (1.45 g, 7.55 mmol) in DMF (20 mL) was stirred at room temperature for 1 hour. Then [2R-hydroxy-3-
[ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] -1S- (phenylmethyl) propylamine hydrochloride (3.825 g, 7.54 mmol) and N-methylmorpholine (3.80 g) were added and the stirring continued for 18 hours. The DMF was removed in vacuo, the residue was dissolved in dichloromethane (500 mL) , and washed with citric acid (IN, 100 mL) , sodium bicarbonate (100 mL) , brine (200 mL) , dried, filtered, and concentrated to afford 4.69 g (91%) of pure N-[2R- hydroxy-3-[ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- [N- (phenyImethoxy carbonyl) amino] -3 , 3-dimethylbutanamide.
Part F: Preparation of N-[2R-hydroxy-3- [ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -2S- (amino) -3 , 3- dimethylbutanamide«dihydrobromide
Figure imgf000164_0002
A solution of N- [2R-hydroxy-3-[ [ (benzothiazol-6- yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propy1 ] -2S-[N-(phenyImethoxycarbonyl) amino] -3 , 3- dimethyl butanamide (4.69 g, 6.89 mmol) in dichloroethane (200 mL) was treated with HBr (48% in acetic acid, 7.1 mL) , and the reaction mixture was stirred for 2 hours at room temperature. The reaction mixture was concentrated and the residue was washed with diethyl ether several times to afford 4.88 g of the desired dihydrobromide product as a powder: high resolution FAB-MS Calcd for C27H38N40 S2 : 547.2413, found: 547.2429 (M+H).
Part G: Preparation of N-[2R-hydroxy-3-
[ [ (benzothiazol-6-yl) sulfonyl] (2-methylpropyl) amino] - IS- (phenylmethyl) propyl] -2S-[ (chloroacetyl) amino] -3 , 3- dimethylbutanamide
A mixture of N-[2R-hydroxy-3-[ [ (benzothiazol-6- yl) sulfonyl] (2-methylpropyl) amino] -IS- (phenylmethyl) propyl] -2S- (amino) -3 , 3-dimethylbutanamide dihydrobromide (3.5 g, 4.9388 mmol), chloroacetic anhydride (0.929 g, 5.44 mmol) and triethylamine (1.097 g, 10.86 mmol) in dichloromethane (35 mL) was stirred at room temperature for 16 hours. The reaction mixture was washed with citric acid (IN, 30 mL) , sodium bicarbonate (30 mL) , brine (30 mL) , dried, filtered and concentrated to afford 3.0 g of the desired product.
EXAMPLE 59
Following the procedures of the previous Examples, the compounds set forth in Tables 2 through 19 can be prepared.
TABLE 2
Figure imgf000166_0001
Entry R4
1 isobutyl 4-aminophen l
2 isobutyl 3-aminopheny1
3 cyclopentylmethyl phenyl
4 cyclohexylmethyl phenyl
5 cyclopentylmethyl 1 , 3-benzodioxol-5-yl
6 c c1ohexylmethy1 1 , 3-benzodioxol-5-yl
7 cyclopentylmethyl benzofuran-5-yl
8 cyclohexylmethyl benzofuran-5-yl
9 cyclopentylmethyl 2 , 3-dihydrobenzofuran-5-yl
10 cyclohexylmethyl 2 , 3-dihydrobenzofuran-5-yl
11 isobutyl 1 , 3-benzodioxol-5-yl
12 isobutyl benzofuran-5-yl
13 isobutyl 2 , 3-dihydrobenzofuran-5-yl
14 isobutyl 1 , 4-benzodioxan-6-yl
15 isoamyl 1, 3-benzodioxol-5-yl
16 isoamyl 2 , 3-dihydrobenzofuran-5-yl
17 isoamyl 1 , 4-benzodioxan-6-yl
18 isobutyl benzothiazol-6-yl
19 isobutyl 2-amino-benzothiazol-6-yl
20 isobutyl benzoxazol-5-yl
21 cyclopentylmethyl 4-methoxypheny1
22 cyclohexylmethyl 4-methoxyphenyl TABLE 3A
Figure imgf000167_0001
Entry
Figure imgf000167_0002
TABLE 3B
Figure imgf000168_0001
Entry
Figure imgf000168_0002
TABLE 4A
Figure imgf000169_0001
Entry R2
(CH3)2CHCH2- (4-FC6H5)CH2
CH3CH2CH2CH2- (naphth-2-yl)CH2
CH3SCH2CH2- * C5HιιCH2
C6H5CH2- C6H5SCH2
(4-CH3OC6H5)CH2- (naphth-2-yl)SCH
TABLE 4B
Figure imgf000169_0002
Entry R2
(CH3)2CHCH2- (4-FC6H5)CH2-
CH3CH2CH2CH2- (naphth-2-yl)CH2-
CH3SCH2CH2- C6HnCH2-
C6H5CH2- C6H5SCH2-
(4-CH3OC6H5)CH2- (naphth-2-yl) SCH2- TABLE 5A
Figure imgf000170_0001
TABLE 5B
Figure imgf000170_0002
TABLE 6
Figure imgf000171_0001
Entry 3
Figure imgf000171_0002
Figure imgf000172_0001
Entry <
Figure imgf000172_0002
Figure imgf000173_0001
Entry R<
Figure imgf000173_0002
TABLE 7C
Figure imgf000174_0001
Entry R-
Figure imgf000174_0002
TABLE 7D
Figure imgf000175_0001
Entry
Figure imgf000175_0002
TABLE 7E
Figure imgf000176_0001
Entry
Figure imgf000176_0002
TABLE 7F
Figure imgf000177_0001
Entry <
Figure imgf000177_0002
TABLE 8
Figure imgf000178_0001
Entry RJ
Figure imgf000178_0002
CH3- CH3\ J
X CH3 : CH3- o o 0 o
Figure imgf000178_0003
TABLE 9
Figure imgf000179_0001
Figure imgf000180_0001
Entry <
Figure imgf000180_0002
Figure imgf000181_0001
Entry
Figure imgf000181_0002
r—NHCOCH2CH3
— NHCONH2
Figure imgf000181_0003
Figure imgf000181_0004
TABLE IOC
Figure imgf000182_0001
Entry 4
Figure imgf000182_0002
NHCOCH2CH3
NHCONH2
Figure imgf000182_0003
Figure imgf000182_0004
Figure imgf000182_0005
Figure imgf000182_0006
TABLE 10D
Figure imgf000183_0001
Entry
Figure imgf000183_0002
Figure imgf000184_0001
Entry 4
Figure imgf000184_0002
Figure imgf000184_0003
Figure imgf000184_0004
Figure imgf000185_0001
Entry
Figure imgf000185_0002
NHCONH2
Figure imgf000185_0003
Figure imgf000185_0004
Figure imgf000185_0005
TABLE 11A
Figure imgf000186_0001
Entry Rv
Figure imgf000186_0002
TABLE 11B
Figure imgf000187_0001
Entry R*
H H
Figure imgf000187_0002
Figure imgf000188_0001
Entry R
H H
Figure imgf000188_0002
Figure imgf000189_0001
Entry R'
Figure imgf000189_0002
TABLE HE
Figure imgf000190_0001
Entry
Figure imgf000190_0002
TABLE 11F
Figure imgf000191_0001
Entry R*
Figure imgf000191_0002
TABLE 12A
Figure imgf000192_0001
Entry
Figure imgf000192_0002
TABLE 12B
Figure imgf000193_0001
Entry
Figure imgf000193_0002
TABLE 13 A
Figure imgf000194_0001
Entry
Figure imgf000194_0002
TABLE 13B
Figure imgf000195_0001
Entry
Figure imgf000195_0002
TABLE 14A
Figure imgf000196_0001
Entry
Figure imgf000196_0002
TABLE 14B
Figure imgf000197_0001
Entry
Figure imgf000197_0002
OH
Figure imgf000197_0003
TABLE 15
Figure imgf000198_0001
Figure imgf000198_0002
Figure imgf000198_0003
TABLE 16A
Figure imgf000199_0001
Entry
Figure imgf000199_0002
CH3OCH2CH20
Figure imgf000199_0003
Figure imgf000199_0004
TABLE 16B
Figure imgf000200_0001
Entry
OH
/
Figure imgf000200_0003
Figure imgf000200_0002
Figure imgf000200_0004
TABLE 17A
Figure imgf000201_0001
Entry
(CH3)2CHCH2- (4-FC6H5)CH2-
CH3CH2CH2CH2- (naphth-2-yl)CH2-
Figure imgf000201_0002
C6H5CH2- C6H5SCH2-
(4-CH3OC6H5)CH2- (naphth-2-yl) SCH2-
TABLE 17B
Figure imgf000201_0003
Entry R'
(CH3)2CHCH2- (4-FC6H5)CH2-
CH3CH2CH2CH2- (naphth-2-yl)CH2-
CH3SCH2CH2- C6HnCH2-
C6H5CH2- C6H5SCH2-
(4-CH3OC6H5)CH2- (naphth-2-yl) SCH2- TABLE 18A
Figure imgf000202_0001
Entry
Figure imgf000202_0002
TABLE 18B
Figure imgf000202_0003
Entry R-
Figure imgf000202_0004
TABLE 19
Figure imgf000203_0001
Entry
Figure imgf000203_0002
EXAMPLE 60
The compounds of the present invention are effective HIV protease inhibitors. Utilizing an enzyme assay as described below, the compounds set forth in the examples herein disclosed inhibited the HIV enzyme. The preferred compounds of the present invention and their calculated IC50 (inhibiting concentration 50%, i.e., the concentration at which the inhibitor compound reduces enzyme activity by 50%) values are shown in Tables 20 and 21. The enzyme method is described below. The substrates is 2-Ila-Nle-Pha (p-N02) ) Gln-ArgJH2. The positive control is MVT-101 (Miller, M. et al, Science, 246, 1149 (1989)] The assay conditions are as follows: Assay buffer: 20 M sodium phosphate, pH 6.4 20% glycerol 1 mM EDTA
1 mM DTT 0.1% CHAPS The above described substrate is dissolved in DMSO, then diluted 10 fold in assay buffer. Final substrate concentration in the assay is 80 μM. HIV protease is diluted in the assay buffer to a final enzyme concentration of 12.3 nanomolar, based on a molecular weight of 10,780.
The final concentration of DMSO is 14% and the final concentration of glycerol is 18%. The test compound is dissolved in DMSO and diluted in DMSO to lOx the test concentration; lOμl of the enzyme preparation is added, the materials mixed and then the mixture is incubated at ambient temperature for 15 minutes. The enzyme reaction is initiated by the addition of 40μl of substrate. The increase in fluorescence is monitored at 4 time points (0, 8, 16 and 24 minutes) at ambient temperature. Each assay is carried out in duplicate wells.
The preceding examples can be repeated with similar success by substituting the generically or specifically described reactants and/or operating conditions of this invention for those used in the preceding examples. TABLE 20
Entry Compound IC50(nM)
Figure imgf000205_0001
EXAMPLE 61
The effectiveness of various compounds were determined in the above-described enzyme assay and in a CEM cell assay. The HIV inhibition assay method of acutely infected cells is an automated tetrazolium based colorimetric assay essentially that reported by Pauwles et al, J. Virol. Methods, 20, 309-321 (1988) . Assays were performed in 96-well tissue culture plates. CEM cells, a CD4+ cell line, were grown in RPMI-1640 medium (Gibco) supplemented with a 10% fetal calf serum and were then treated with polybrene (2μg/ml) . An 80 μl volume of medium containing 1 x 10 cells was dispensed into each well of the tissue culture plate. To each well was added a lOOμl volume of test compound dissolved in tissue culture medium (or medium without test compound as a control) to achieve the desired final concentration and the cells were incubated at 37 °C for 1 hour. A frozen culture of HIV-1 was diluted in culture medium to a concentration of 5 x 104 TCID50 per ml (TCID50 = the dose of virus that infects 50% of cells in tissue culture) , and a 20μL volume of the virus sample (containing 1000 TCID50 of virus) was added to wells containing test compound and to wells containing only medium (infected control cells) . Several wells received culture medium without virus (uninfected control cells) . Likewise, the intrinsic toxicity of the test compound was determined by adding medium without virus to several wells containing test compound. In summary, the tissue culture plates contained the following experiments:
Cells Drug Virus
1 . +
2 . + +
3 . + + 4 4.. + + + +
In experiments 2 and 4 the final concentrations of test compounds were 1, 10, 100 and 500 μg/ l. Either azidothymidine (AZT) or dideoxyinosine (ddl) was included as a positive drug control. Test compounds were dissolved in DMSO and diluted into tissue culture medium so that the final DMSO concentration did not exceed 1.5% in any case. DMSO was added to all control wells at an appropriate concentration.
Following the addition of virus, cells were incubated at 37 °C in a humidified, 5% C02 atmosphere for 7 days. Test compounds could be added on days 0, 2 and 5 if desired. On day 7, post-infection, the cells in each well were resuspended and a lOOμl sample of each cell suspension was removed for assay. A 20μL volume of a 5 mg/ml solution of 3-(4,5-dimethylthiazol-2-yl) -2,5- diphenyltetrazolium bromide (MTT) was added to each lOOμL cell suspension, and the cells were incubated for 4 hours at 27°C in a 5% C02 environment. During this incubation, MTT is metabolically reduced by living cells resulting in the production in the cell of a colored formazan product. To each sample was added lOOμl of 10% sodium dodecylsulfate in 4.01 N HG1 to lyse the cells, and samples were incubated overnight. The absorbance at 590 n was determined for each sample using a Molecular Devices icroplate reader. Absorbance values for each set of wells is compared to assess viral control infection, uninfected control cell response as well as test compound by cytotoxicity and antiviral efficacy.
TABLE 21
Entry Compound IC50 EC50 (nM) (nM)
Figure imgf000207_0001
TABLE 21 (cont'd)
Entry Compound IC50 EC50 (nM) (nM)
Figure imgf000208_0001
TABLE 21 (cont'd)
Entry Compound IC50 EC50 (nM) (nM)
Figure imgf000209_0001
TABLE 21 (cont'd)
Entry Compound IC50 EC50 (nM) (nM)
Figure imgf000210_0001
The compounds of the present invention are effective antiviral compounds and, in particular, are effective retroviral inhibitors as shown above. Thus, the subject compounds are effective HIV protease inhibitors. It is contemplated that the subject compounds will also inhibit other retroviruses such as other lentiviruses in particular other strains of HIV, e.g. HIV-2 , human T-cell leukemia virus, respiratory syncitial virus, simia immunodeficiency virus, feline leukemia virus, feline immuno-deficiency virus, hepadnavirus, cytomegalovirus and picornavirus. Thus, the subject compounds are effective in the treatment, proplylaxis of retroviral infections and/or the prevention of the spread of retroviral infections.
The subject compounds are also effective in preventing the growth of retroviruses in a solution. Both human and animal cell cultures, such as T- lymphocyte cultures, are utilized for a variety of well known purposes, such as research and diagnostic procedures including calibrators and controls. Prior to and during the growth and storage of a cell culture, the subject compounds may be added to the cell culture medium at an effective concentration to prevent the unexpected or undesired replication of a retrovirus that may inadvertently, unknowingly or knowingly be present in the cell culture. The virus may be present originally in the cell culture, for example HIV is known to be present in human T-lymphocytes long before it is detectable in blood, or through exposure to the virus. This use of the subject compounds prevents the unknowing or inadvertent exposure of a potentially lethal retrovirus to a researcher or clinician. Compounds of the present invention can possess one or more asymmetric carbon atoms and are thus capable of existing in the form of optical isomers as well as in the form of racemic or nonracemic mixtures thereof. The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example by formation of diastereoisomeric salts by treatment with an optically active acid or base. Examples of appropriate acids are tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric and camphorsulfonic acid and then separation of the mixture of diastereoisomers by crystallization followed by liberation of the optically active bases from these salts. A different process for separation of optical isomers involves the use of a chiral chromatography column optimally chosen to maximize the separation of the enantiomers. Still another available method involves synthesis of covalent diastereoisomeric molecules by reacting compounds of Formula I with an optically pure acid in an activated form or an optically pure isocyanate. The synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound. The optically active compounds of Formula I can likewise be obtained by utilizing optically active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt.
The compounds of the present invention can be used in the form of salts derived from inorganic or organic acids. These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobro ide, hydroiodide, 2-hydroxy-ethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2- naphthalenesulfonate, oxalate, palmoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, mesylate and undecanoate. Also, the basic nitrogen- containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl"bromides, and others. Water or oil-soluble or dispersible products are thereby obtained.
Examples of acids which may be employed to form pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, sulphuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, succinic acid and citric acid. Other examples include salts with alkali metals or alkaline earth metals, such as sodium, potassium, calcium or magnesium or with organic bases.
Total daily dose administered to a host in single or divided doses may be in amounts, for example, from 0.001 to 10 mg/kg body weight daily and more usually 0.01 to 1 mg. Dosage unit compositions may contain such amounts of submultiples thereof to make up the daily dose.
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.
The dosage regimen for treating a disease condition with the compounds and/or compositions of this invention is selected in accordance with a variety of factors, including the type, age, weight, sex, diet and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetic and toxicology profiles^ of the particular compound employed, whether a drug delivery system is utilized and whether the compound is administered as part of a drug combination. Thus, the dosage regimen actually employed may vary widely and therefore may deviate from the preferred dosage regimen set forth above.
The compounds of the present invention may be administered orally, parenterally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. Topical administration may also involve the use of transdermal administration such as transdermal patches or iontophoresis devices. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection, or infusion techniques. Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol . Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic 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. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable nonirritating excipient such as cocoa butter and polyethylene glycols which are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound may be admixed with at least one inert diluent such as sucrose lactose or starch. Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g. , lubricating agents such as magnesium stearate. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring, and perfuming agents. While the compounds of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more immunomodulatorε, antiviral agents or other antiinfective agents. For example, the compounds of the invention can be administered in combination with AZT, DDI, DDC or with glucosidase inhibitors, such as N-butyl-1-deoxynojirimycin or prodrugs thereof, for the prophylaxis and/or treatment of AIDS. When administered as a combination, the therapeutic agents can be formulated as separate compositions which are given at the same time or different times, or the therapeutic agents can be given as a single composition.
The foregoing is merely illustrative of the invention and is not intended to limit the invention to the disclosed compounds. Variations and changes which are obvious to one skilled in the art are intended to be within the scope and nature of the invention which are defined in the appended claims. From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.

Claims

WHAT IS CLAIMED IS:
A process for preparing a benzo fused heterocyclo sulfonyl halide comprising reacting a benzo fused heterocyclic compound with an S03 complex in the presence of a water immiscible, non-reactive solvent, at a temperature of from about 0°C to about 75°C, cooling, if necessary, to a temperature of from about -25°C to about 65°C and then adding an oxalyl halide.
2. The process of claim 1 wherein the S03 complex is S03DMF.
The process of claim 1 wherein the water immiscible solvent is dichloroethane, methylene chloride, trichloroethane, toluene, tetrachloroethane, or mixtures thereof.
4. The process of claim 3 wherein the water immiscible solvent is dichloroethane.
The process of claim 1 wherein the oxalyl halide is oxalyl chloride.
6. The process of claim 1 wherein the benzo fused heterocyclo sulfonyl halide is benzo fused heterocyclo sulfonyl chloride. The process of claim 1 wherein the benzo fused heterocyclo compound has the formula:
Figure imgf000218_0001
wherein A and B each independently represent 0 or s;
R represents a hydrogen, deuterium, alkyl or halogen radical; and
R7 represents a hydrogen, deuterium, alkyl or halogen radical;
8. The process of claim 7 wherein A and B each represent 0; R6 represents a hydrogen, deuterium, alkyl of 1-5 carbon atoms, fluoro or chloro radical; and R7 represents hydrogen, deuterium, alkyl of 1-3 carbon atoms, fluoro or chloro radical.
The process of claim 8 wherein R6 represents a hydrogen, deuterium, methyl, ethyl, propyl, isopropyl or fluoro radical; and R7 represents a hydrogen, deuterium, methyl or fluoro radical.
10. The process of claim 7 wherein A and B represent O; and R and R each independently represent a fluoro or chloro radical.
11. The process of claim 10 wherein R6 and R7 each represent a fluoro radical.
12. The process of claim 7 wherein R6 and R7 each represent a hydrogen.
13. The process of claim 7 wherein A and B represent 0; and R6 and R7 each represent a hydrogen.
14. The process of claim 1 wherein the benzo fused heterocyclic compound is a benzodioxane, benzopyran, or benzodihydrofuran.
15. The process of claim 14 wherein the benzo fused heterocyclic compound is 1,4-benzodioxane, 2,3- benzodihydropyran, or 2, 3-benzodihydrofuran.
16. The process of claim 1 comprising a single pot process .
PCT/US1999/007047 1998-05-19 1999-05-18 Process for the preparation of a benzo fused heterocyclo sulfonyl halide intermediate WO1999059989A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU38604/99A AU3860499A (en) 1998-05-19 1999-05-18 Process for the preparation of a benzo fused heterocyclo sulfonyl halide intermediate

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/080,928 US6140505A (en) 1995-03-10 1998-05-19 Synthesis of benzo fused heterocyclic sulfonyl chlorides
US09/080,928 1998-05-19

Publications (1)

Publication Number Publication Date
WO1999059989A1 true WO1999059989A1 (en) 1999-11-25

Family

ID=22160564

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/007047 WO1999059989A1 (en) 1998-05-19 1999-05-18 Process for the preparation of a benzo fused heterocyclo sulfonyl halide intermediate

Country Status (3)

Country Link
US (6) US6140505A (en)
AU (1) AU3860499A (en)
WO (1) WO1999059989A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003076413A1 (en) * 2002-03-12 2003-09-18 Tibotec Pharmaceuticals Ltd Broadspectrum substituted benzimidazole sulfonamide hiv protease inhibitors

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6140505A (en) 1995-03-10 2000-10-31 G. D. Searle & Co. Synthesis of benzo fused heterocyclic sulfonyl chlorides
EE05385B1 (en) * 2001-02-14 2011-02-15 Tibotec�Pharmaceuticals�Ltd. Broad-spectrum Á2- (Substituted Áamino) Benzothiazole Sulphonamides ÁkuiÁHIVÁProtease Inhibitors, Methods of Preparation, and UseUnited Pharmaceutical Formulations
BRPI0821247A2 (en) * 2007-12-14 2015-06-16 Univ Georgetown Histone Deacetylase Inhibitors
WO2009129532A1 (en) * 2008-04-18 2009-10-22 University Of Connecticut Compounds for lysosomal modulation and methods of use
PE20140406A1 (en) 2010-11-18 2014-04-16 Univ Yale BIFUNCTIONAL MOLECULES WITH ANTIBODY RECRUITMENT ACTIVITY AND INITIAL INHIBITORY ACTIVITY AGAINST HUMAN IMMUNODEFICIENCY VIRUS
CN102108069B (en) * 2011-01-07 2012-09-26 上海优贝德生物医药有限公司 Method for preparing 6-benzothiazole sulfonyl chloride
US20120309796A1 (en) 2011-06-06 2012-12-06 Fariborz Firooznia Benzocycloheptene acetic acids
US20160075643A1 (en) * 2014-09-16 2016-03-17 Zcl Chemicals Limited Novel process to prepare intermediates of hiv-protease inhibitors thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0583960A2 (en) * 1992-08-19 1994-02-23 Eli Lilly And Company Synthesis of bicyclic aromatic sulfonic acids, sulfonyl chlorides and sulfonamides
WO1996022287A1 (en) * 1995-01-20 1996-07-25 G.D. Searle & Co. Bis-sulfonamide hydroxyethylamino retroviral protease inhibitors
WO1996028418A1 (en) * 1995-03-10 1996-09-19 G.D. Searle & Co. Sulfonylalkanoylamino hydroxyethylamino sulfonamide retroviral protease inhibitors
WO1997018205A1 (en) * 1995-11-15 1997-05-22 G.D. Searle & Co. Substituted sulfonylalkanoylamino hydroxyethylamino sulfonamide retroviral protease inhibitors

Family Cites Families (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5142056A (en) 1989-05-23 1992-08-25 Abbott Laboratories Retroviral protease inhibiting compounds
DE3269604D1 (en) 1981-06-26 1986-04-10 Schering Corp Novel imidazo(1,2-a)pyridines and pyrazines, processes for their preparation and pharmaceutical compositions containing them
US4634465A (en) 1982-07-16 1987-01-06 Ciba-Geigy Corporation Fused N-phenylsulfonyl-N'-pyrimidinylureas and N-phenylsulfonyl-N'triazinylureas
EP0104041B1 (en) 1982-09-15 1988-07-27 Aktiebolaget Hässle Enzyme inhibitors
US4595407A (en) 1982-11-01 1986-06-17 E. I. Du Pont De Nemours And Company Triazinyl-amino-carbonyl-1,3-benzohetero- or -1,4-benzohetero-sulfonamides
EP0114993B1 (en) 1982-12-27 1990-05-16 Merck & Co. Inc. Renin inhibitory tripeptides
US4668770A (en) 1982-12-27 1987-05-26 Merck & Co., Inc. Renin inhibitory tripeptides
WO1984003044A1 (en) 1983-02-07 1984-08-16 Ferring Ab Enzyme inhibitors
JPS59227851A (en) 1983-06-09 1984-12-21 Sankyo Co Ltd Peptides having inhibitory action on renin
US4514391A (en) 1983-07-21 1985-04-30 E. R. Squibb & Sons, Inc. Hydroxy substituted peptide compounds
US4477441A (en) 1983-09-14 1984-10-16 Merck & Co., Inc. Renin inhibitors containing a C-terminal disulfide cycle
US4645759A (en) 1984-06-22 1987-02-24 Abbott Laboratories Renin inhibiting compounds
US4616088A (en) 1984-10-29 1986-10-07 E. R. Squibb & Sons, Inc. Amino acid ester and amide renin inhibitor
US4668769A (en) 1985-08-02 1987-05-26 Hoover Dennis J Oxa- and azahomocyclostatine polypeptides
US4599198A (en) 1985-08-02 1986-07-08 Pfizer Inc. Intermediates in polypeptide synthesis
ES8604955A1 (en) 1985-11-08 1986-03-16 Covex Sa Process for the preparation of (-) eburnamenin-14-(15H)-one resinates
CA1282549C (en) 1985-11-12 1991-04-02 Eric M. Gordon Aminocarbonyl renin inhibitors
US4757050A (en) 1985-12-23 1988-07-12 E. R. Squibb Sons, Inc. Ureido renin inhibitors
CA1297631C (en) 1985-12-23 1992-03-17 Sesha I. Natarajan Ureido renin inhibitors
US4880938A (en) 1986-06-16 1989-11-14 Merck & Co., Inc. Amino acid analogs
IL83215A (en) * 1986-07-23 1991-12-15 Lilly Co Eli Synthesis of bicyclic heteroaromatic sulfonyl chlorides
DE3635907A1 (en) 1986-10-22 1988-04-28 Merck Patent Gmbh HYDROXY AMINO ACID DERIVATIVES
CH676988A5 (en) 1987-01-21 1991-03-28 Sandoz Ag
USH725H (en) 1987-02-26 1990-01-02 E. R. Squibb & Sons, Inc. Ureido amino and imino acids, compositions and methods for use
GB8707412D0 (en) 1987-03-27 1987-04-29 Fujisawa Pharmaceutical Co Peptide compounds
DE3830825A1 (en) 1987-09-15 1989-03-23 Sandoz Ag Hydrophilic renin inhibitors, their preparation and use
IL89900A0 (en) 1988-04-12 1989-12-15 Merck & Co Inc Hiv protease inhibitors useful for the treatment of aids and pharmaceutical compositions containing them
US4977277A (en) 1988-05-09 1990-12-11 Abbott Laboratories Functionalized peptidyl aminodiols and -triols 4-amino-5-cyclohexyl-3-hydroxy-1,2-oxopentane and derivatives thereof
IL90218A0 (en) 1988-05-13 1989-12-15 Abbott Lab Retroviral protease inhibitors
CA1340588C (en) 1988-06-13 1999-06-08 Balraj Krishan Handa Amino acid derivatives
IL91307A0 (en) 1988-08-24 1990-03-19 Merck & Co Inc Hiv protease inhibitors and pharmaceutical compositions for the treatment of aids containing them
CA2012306A1 (en) 1989-03-28 1990-09-28 Werner Neidhart Amino acid derivatives
JP2701932B2 (en) 1989-04-10 1998-01-21 サントリー株式会社 Protease inhibitor
DE3912829A1 (en) 1989-04-19 1990-10-25 Bayer Ag USE OF RENININHIBITIC PEPTIDES AS AGENTS AGAINST RETROVIRUS
TW225540B (en) 1990-06-28 1994-06-21 Shionogi & Co
US5289728A (en) 1990-11-08 1994-03-01 Jr Johanson, Inc. Flow-no-flow tester
EP0558673B1 (en) 1990-11-19 1996-04-17 Monsanto Company Retroviral protease inhibitors
ZA929869B (en) 1991-12-20 1994-06-20 Syntex Inc Hiv protease inhibitors
JP3657002B2 (en) 1992-08-25 2005-06-08 ジー.ディー.サール アンド カンパニー Alpha- and beta-amino acid hydroxyethylaminosulfonamides useful as retroviral protease inhibitors
IS2334B (en) 1992-09-08 2008-02-15 Vertex Pharmaceuticals Inc., (A Massachusetts Corporation) Aspartyl protease inhibitor of a new class of sulfonamides
US6140505A (en) * 1995-03-10 2000-10-31 G. D. Searle & Co. Synthesis of benzo fused heterocyclic sulfonyl chlorides
US5756533A (en) * 1995-03-10 1998-05-26 G.D. Searle & Co. Amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
JP3680749B2 (en) * 2001-03-23 2005-08-10 ヤマハ株式会社 Automatic composer and automatic composition program

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0583960A2 (en) * 1992-08-19 1994-02-23 Eli Lilly And Company Synthesis of bicyclic aromatic sulfonic acids, sulfonyl chlorides and sulfonamides
WO1996022287A1 (en) * 1995-01-20 1996-07-25 G.D. Searle & Co. Bis-sulfonamide hydroxyethylamino retroviral protease inhibitors
WO1996028418A1 (en) * 1995-03-10 1996-09-19 G.D. Searle & Co. Sulfonylalkanoylamino hydroxyethylamino sulfonamide retroviral protease inhibitors
WO1997018205A1 (en) * 1995-11-15 1997-05-22 G.D. Searle & Co. Substituted sulfonylalkanoylamino hydroxyethylamino sulfonamide retroviral protease inhibitors

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003076413A1 (en) * 2002-03-12 2003-09-18 Tibotec Pharmaceuticals Ltd Broadspectrum substituted benzimidazole sulfonamide hiv protease inhibitors
EA011946B1 (en) * 2002-03-12 2009-06-30 Тиботек Фармасьютикалз Лтд. Broad spectrum substituted benzimidazole sulfonamide hiv protease inhibitors
AP2111A (en) * 2002-03-12 2010-03-03 Tibotec Pharm Ltd Broadspectrum substituted benzimidazole sulfonamide HIV protease inhibitors.
US8143421B2 (en) 2002-03-12 2012-03-27 Tibotec Pharmaceuticals Ltd. Broadspectrum substituted benzimidazole sulfonamide HIV protease inhibitors
HRP20040936B1 (en) * 2002-03-12 2012-09-30 Tibotec Pharmaceuticals Ltd. Broadspectrum substituted benzimidazole sulfonamide hiv protease inhibitors

Also Published As

Publication number Publication date
US6310080B1 (en) 2001-10-30
US20020111368A1 (en) 2002-08-15
US20040260095A1 (en) 2004-12-23
US20060189611A1 (en) 2006-08-24
AU3860499A (en) 1999-12-06
US6730669B2 (en) 2004-05-04
US6458785B1 (en) 2002-10-01
US7098202B2 (en) 2006-08-29
US6140505A (en) 2000-10-31
US20030216435A1 (en) 2003-11-20

Similar Documents

Publication Publication Date Title
EP0813542B1 (en) Amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US6316496B1 (en) Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
EP1052250B1 (en) Sulfonylalkanoylamino hydroxyethylamino sulfonamide retroviral protease inhibitors
EP0815124B1 (en) Heterocyclecarbonyl amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
WO1997018205A1 (en) Substituted sulfonylalkanoylamino hydroxyethylamino sulfonamide retroviral protease inhibitors
US20060189611A1 (en) Amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US6172101B1 (en) Heterocyclecarbonyl amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US6683210B2 (en) Bis-amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors
US6407134B1 (en) Heterocyclecarbonyl amino acid hydroxyethylamino sulfonamide retroviral protease inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID 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 SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

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

Ref country code: KR

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase